1
|
Kim KH, Lee SJ, Kim J, Moon Y. Leveraging Xenobiotic-Responsive Cancer Stemness in Cell Line-Based Tumoroids for Evaluating Chemoresistance: A Proof-of-Concept Study on Environmental Susceptibility. Int J Mol Sci 2024; 25:11383. [PMID: 39518936 PMCID: PMC11545740 DOI: 10.3390/ijms252111383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 10/16/2024] [Accepted: 10/20/2024] [Indexed: 11/16/2024] Open
Abstract
Emerging evidence suggests that cancer stemness plays a crucial role in tumor progression, metastasis, and chemoresistance. Upon exposure to internal or external stress, ribosomes stand sentinel and facilitate diverse biological processes, including oncological responses. In the present study, ribosome-inactivating stress (RIS) was evaluated for its modulation of cancer cell stemness as a pivotal factor of tumor cell reprogramming. Based on the concept of stress-responsive cancer cell stemness, we addressed human intestinal cancer cell line-based off-the-shelf spheroid cultures. Intestinal cancer cell line-based spheroids exhibited heightened levels of CD44+CD133+ cancer stemness, which was improved by chemical-induced RIS. Further evaluations revealed the potential of these stress-imprinted spheroids as a platform for chemoresistance screening. Compared to adherent cells, stemness-improved spheroid cultures displayed reduced apoptosis in response to 5-fluorouracil (5-FU), a frontline chemotherapeutic agent against colorectal cancer. Moreover, serial subcultures with repeated RIS exposure maintained and even enhanced cancer stemness and chemoresistance patterns. In particular, isolated CD44+CD133+ cancer stem cells exhibited higher chemoresistance compared to unsorted cells. To elucidate the mechanisms underlying RIS-induced stemness, RNA-seq analysis identified Wnt signaling pathways and stemness-associated signals as notable features in spheroids exposed to RIS. Loss-of-function studies targeting connective tissue growth factor (CTGF), a negative regulator of Wnt signaling, revealed that CTGF-deficient spheroids exhibited improved cancer stemness and resistance to 5-FU, with RIS further enhancing these effects. In conclusion, this proof-of-concept study demonstrates the feasibility of leveraging stress-responsive cancer stemness for the development of spheroid-based platforms for chemoresistance evaluation and elucidation of pathophysiological processes of colorectal tumorigenesis under environmental stress.
Collapse
Affiliation(s)
- Ki-Hyung Kim
- Laboratory of Mucosal Exposome and Biomodulation, Department of Integrative Biomedical Sciences, Pusan National University, Yangsan 50612, Republic of Korea; (K.-H.K.); (S.J.L.); (J.K.)
- Department of Obstetrics and Gynecology, College of Medicine, Pusan National University, Busan 49241, Republic of Korea
- Biomedical Research Institute, Pusan National University, Busan 49241, Republic of Korea
| | - Seung Joon Lee
- Laboratory of Mucosal Exposome and Biomodulation, Department of Integrative Biomedical Sciences, Pusan National University, Yangsan 50612, Republic of Korea; (K.-H.K.); (S.J.L.); (J.K.)
| | - Juil Kim
- Laboratory of Mucosal Exposome and Biomodulation, Department of Integrative Biomedical Sciences, Pusan National University, Yangsan 50612, Republic of Korea; (K.-H.K.); (S.J.L.); (J.K.)
| | - Yuseok Moon
- Laboratory of Mucosal Exposome and Biomodulation, Department of Integrative Biomedical Sciences, Pusan National University, Yangsan 50612, Republic of Korea; (K.-H.K.); (S.J.L.); (J.K.)
- Biomedical Research Institute, Pusan National University, Busan 49241, Republic of Korea
- Graduate Program of Genomic Data Sciences, Pusan National University, Yangsan 50612, Republic of Korea
| |
Collapse
|
2
|
McCoy R, Oldroyd S, Yang W, Wang K, Hoven D, Bulmer D, Zilbauer M, Owens RM. In Vitro Models for Investigating Intestinal Host-Pathogen Interactions. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306727. [PMID: 38155358 PMCID: PMC10885678 DOI: 10.1002/advs.202306727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 12/01/2023] [Indexed: 12/30/2023]
Abstract
Infectious diseases are increasingly recognized as a major threat worldwide due to the rise of antimicrobial resistance and the emergence of novel pathogens. In vitro models that can adequately mimic in vivo gastrointestinal physiology are in high demand to elucidate mechanisms behind pathogen infectivity, and to aid the design of effective preventive and therapeutic interventions. There exists a trade-off between simple and high throughput models and those that are more complex and physiologically relevant. The complexity of the model used shall be guided by the biological question to be addressed. This review provides an overview of the structure and function of the intestine and the models that are developed to emulate this. Conventional models are discussed in addition to emerging models which employ engineering principles to equip them with necessary advanced monitoring capabilities for intestinal host-pathogen interrogation. Limitations of current models and future perspectives on the field are presented.
Collapse
Affiliation(s)
- Reece McCoy
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgeCambridgeCB3 0ASUK
| | - Sophie Oldroyd
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgeCambridgeCB3 0ASUK
| | - Woojin Yang
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgeCambridgeCB3 0ASUK
- Wellcome‐MRC Cambridge Stem Cell InstituteUniversity of CambridgeCambridgeCB2 0AWUK
| | - Kaixin Wang
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgeCambridgeCB3 0ASUK
| | - Darius Hoven
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgeCambridgeCB3 0ASUK
| | - David Bulmer
- Department of PharmacologyUniversity of CambridgeCambridgeCB2 1PDUK
| | - Matthias Zilbauer
- Wellcome‐MRC Cambridge Stem Cell InstituteUniversity of CambridgeCambridgeCB2 0AWUK
| | - Róisín M. Owens
- Department of Chemical Engineering and BiotechnologyUniversity of CambridgeCambridgeCB3 0ASUK
| |
Collapse
|
3
|
Kim J, Jeong H, Ray N, Kim KH, Moon Y. Gut ribotoxic stress responses facilitate dyslipidemia via metabolic reprogramming: an environmental health prediction. Theranostics 2024; 14:1289-1311. [PMID: 38323314 PMCID: PMC10845207 DOI: 10.7150/thno.88586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 01/05/2024] [Indexed: 02/08/2024] Open
Abstract
Rationale: The gut and its accessory organ, the liver, are crucial determinants of metabolic homeostasis via the regulation of circulating lipids for cardiovascular health. In response to environmental insults, cells undergo diverse adaptation or pathophysiological processes via stress-responsive eukaryotic initiation factor 2 alpha (eIF2α) kinase signaling and subsequent cellular reprogramming. We noted that patients with inflammatory gut distress display enhanced levels of ribosomal stress-responsive eIF2α kinase, which is notably associated with lipid metabolic process genes. Based on an assumption that eukaryotic ribosomes are a promising stress-responsive module for molecular reprogramming, chemical ribosome-inactivating stressors (RIS) were assessed for their involvement in enterohepatic lipid regulation. Methods: Experimental assessment was based on prediction using the clinical transcriptome and single-cell RNA-sequencing analysis of inflammatory bowel diseases and obesity. The prediction was verified using RIS exposure models of mice, gut organoids, and intestinal cells. The lipidomic profiling was performed to address RIS-induced intracellular fat alterations. Biochemical processes of the mechanisms were evaluated using RT-PCR, western blot analysis, luciferase reporter assays, and confocal microscopy of genetically ablated or chemically inhibited mice, organoids, and cells. Results: Chemical RIS including deoxynivalenol promoted enterohepatic lipid sequestration while lowering blood LDL cholesterol in normal and diet-induced obese mice. Although ribosomal stress caused extensive alterations in cellular lipids and metabolic genes, the cholesterol import-associated pathway was notably modulated. In particular, ribosomal stress enhanced gut levels of the low-density lipoprotein receptor (LDLR) via both transcriptional and post-transcriptional regulation. Subsequently, LDLR facilitated enterohepatic cholesterol accumulation, leading to dyslipidemia in response to ribosomal stress. Moreover, genetic features of stress-responsive LDLR modulators were consistently proven in the inflammation- and obesity-associated gut model. Conclusion: The elucidated ribosome-linked gut lipid regulation provides predictive insights into stress-responsive metabolic rewiring in chronic human diseases as an environmental health prediction.
Collapse
Affiliation(s)
- Juil Kim
- Laboratory of Mucosal Exposome and Biomodulation, Department of Integrative Biomedical Sciences, Pusan National University, Yangsan, Korea
| | - Hoyoung Jeong
- Laboratory of Mucosal Exposome and Biomodulation, Department of Integrative Biomedical Sciences, Pusan National University, Yangsan, Korea
| | - Navin Ray
- Laboratory of Mucosal Exposome and Biomodulation, Department of Integrative Biomedical Sciences, Pusan National University, Yangsan, Korea
| | - Ki-Hyung Kim
- Laboratory of Mucosal Exposome and Biomodulation, Department of Integrative Biomedical Sciences, Pusan National University, Yangsan, Korea
- Department of Obstetrics and Gynecology, College of Medicine, Pusan National University, Pusan National University, Busan, Korea
- Biomedical Research Institute, Pusan National University, Busan, Korea
| | - Yuseok Moon
- Laboratory of Mucosal Exposome and Biomodulation, Department of Integrative Biomedical Sciences, Pusan National University, Yangsan, Korea
- Biomedical Research Institute, Pusan National University, Busan, Korea
- Graduate Program of Genomic Data Sciences, Pusan National University, Yangsan 50612, Korea
| |
Collapse
|
4
|
Ali M, Xu C, Nawaz S, Ahmed AE, Hina Q, Li K. Anti-Cryptosporidial Drug-Discovery Challenges and Existing Therapeutic Avenues: A "One-Health" Concern. Life (Basel) 2024; 14:80. [PMID: 38255695 PMCID: PMC10820218 DOI: 10.3390/life14010080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 12/20/2023] [Accepted: 12/28/2023] [Indexed: 01/24/2024] Open
Abstract
Cryptosporidiosis is the leading cause of life-threatening diarrheal infection, especially in infants. Oocysts contaminate the environment, and also, being a zoonotic disease, cryptosporidiosis is a threat to One Health. Nitazoxanide is the only FDA-approved drug, effective only in immunocompetent adults, and is not safe for infants. The absence of mitochondria and apicoplast, the presence of an electron-dense band (ED band), hindrances in its genetic and phenotypic manipulations, and its unique position inside the host cell are some challenges to the anti-cryptosporidial drug-discovery process. However, many compounds, including herbal products, have shown efficacy against Cryptosporidium during in vitro and in vivo trials. Still, the "drug of choice" against this protozoan parasite, especially in immunocompromised individuals and infants, has not yet been explored. The One-Health approach addresses this issue, focusing on the intersection of animal, human, and environmental health. The objective of this review is to provide knowledge about novel anti-cryptosporidial drug targets, available treatment options with associated limitations, and possible future shifts toward natural products to treat cryptosporidiosis. The current review is organized to address the treatment and prevention of cryptosporidiosis. An anti-cryptosporidial drug that is effective in immunocompromised individuals and infants is a necessity of our time.
Collapse
Affiliation(s)
- Munwar Ali
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (M.A.); (C.X.)
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Chang Xu
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (M.A.); (C.X.)
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Shah Nawaz
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China;
| | - Ahmed Ezzat Ahmed
- Biology Department, College of Science, King Khalid University, Abha 61413, Saudi Arabia;
| | - Qazal Hina
- Department of Animal Nutrition, University of Veterinary and Animal Sciences, Lahore 54000, Pakistan;
| | - Kun Li
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; (M.A.); (C.X.)
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
5
|
Gunasekera S, Clode PL, King B, Monis P, Thierry B, Carr JM, Chopra A, Watson M, O'Dea M, Hijjawi N, Ryan U. Comparison of in vitro growth characteristics of Cryptosporidium hominis (IdA15G1) and Cryptosporidium parvum (Iowa-IIaA17G2R1 and IIaA18G3R1). Parasitol Res 2023; 122:2891-2905. [PMID: 37776335 PMCID: PMC10667462 DOI: 10.1007/s00436-023-07979-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 09/14/2023] [Indexed: 10/02/2023]
Abstract
Cryptosporidium is a major cause of diarrhoeal disease and mortality in young children in resource-poor countries, for which no vaccines or adequate therapeutic options are available. Infection in humans is primarily caused by two species: C. hominis and C. parvum. Despite C. hominis being the dominant species infecting humans in most countries, very little is known about its growth characteristics and life cycle in vitro, given that the majority of our knowledge of the in vitro development of Cryptosporidium has been based on C. parvum. In the present study, the growth and development of two C. parvum isolates (subtypes Iowa-IIaA17G2R1 and IIaA18G3R1) and one C. hominis isolate (subtype IdA15G1) in HCT-8 cells were examined and compared at 24 h and 48 h using morphological data acquired with scanning electron microscopy. Our data indicated no significant differences in the proportion of meronts or merozoites between species or subtypes at either time-point. Sexual development was observed at the 48-h time-point across both species through observations of both microgamonts and macrogamonts, with a higher frequency of macrogamont observations in C. hominis (IdA15G1) cultures at 48-h post-infection compared to both C. parvum subtypes. This corresponded to differences in the proportion of trophozoites observed at the same time point. No differences in proportion of microgamonts were observed between the three subtypes, which were rarely observed across all cultures. In summary, our data indicate that asexual development of C. hominis is similar to that of C. parvum, while sexual development is accelerated in C. hominis. This study provides new insights into differences in the in vitro growth characteristics of C. hominis when compared to C. parvum, which will facilitate our understanding of the sexual development of both species.
Collapse
Affiliation(s)
- Samantha Gunasekera
- Harry Butler Institute, College of Environmental and Life Sciences, Murdoch University, Murdoch, Western Australia, 6150, Australia.
| | - Peta L Clode
- Centre for Microscopy, Characterisation, and Analysis and School of Biological Sciences, The University of Western Australia, Crawley, Western Australia, 6009, Australia
| | - Brendon King
- South Australian Water Corporation, Adelaide, South Australia, 5000, Australia
| | - Paul Monis
- South Australian Water Corporation, Adelaide, South Australia, 5000, Australia
| | - Benjamin Thierry
- Future Industries Institute, University of South Australia, Adelaide, South Australia, 5095, Australia
| | - Jillian M Carr
- College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, Bedford Park, South Australia, 5042, Australia
| | - Abha Chopra
- Immunology and Infectious Diseases, Murdoch University, Murdoch, Western Australia, 6150, Australia
| | - Mark Watson
- Immunology and Infectious Diseases, Murdoch University, Murdoch, Western Australia, 6150, Australia
| | - Mark O'Dea
- Harry Butler Institute, College of Environmental and Life Sciences, Murdoch University, Murdoch, Western Australia, 6150, Australia
| | - Nawal Hijjawi
- Department of Medical Laboratory Sciences, Faculty of Applied Health Sciences, The Hashemite University, P.O. Box 150459, Zarqa, 13115, Jordan
| | - Una Ryan
- Harry Butler Institute, College of Environmental and Life Sciences, Murdoch University, Murdoch, Western Australia, 6150, Australia.
| |
Collapse
|
6
|
Ren Z, Harriot AD, Mair DB, Chung MK, Lee PHU, Kim DH. Biomanufacturing of 3D Tissue Constructs in Microgravity and their Applications in Human Pathophysiological Studies. Adv Healthc Mater 2023; 12:e2300157. [PMID: 37483106 DOI: 10.1002/adhm.202300157] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 06/27/2023] [Indexed: 07/25/2023]
Abstract
The growing interest in bioengineering in-vivo-like 3D functional tissues has led to novel approaches to the biomanufacturing process as well as expanded applications for these unique tissue constructs. Microgravity, as seen in spaceflight, is a unique environment that may be beneficial to the tissue-engineering process but cannot be completely replicated on Earth. Additionally, the expense and practical challenges of conducting human and animal research in space make bioengineered microphysiological systems an attractive research model. In this review, published research that exploits real and simulated microgravity to improve the biomanufacturing of a wide range of tissue types as well as those studies that use microphysiological systems, such as organ/tissue chips and multicellular organoids, for modeling human diseases in space are summarized. This review discusses real and simulated microgravity platforms and applications in tissue-engineered microphysiological systems across three topics: 1) application of microgravity to improve the biomanufacturing of tissue constructs, 2) use of tissue constructs fabricated in microgravity as models for human diseases on Earth, and 3) investigating the effects of microgravity on human tissues using biofabricated in vitro models. These current achievements represent important progress in understanding the physiological effects of microgravity and exploiting their advantages for tissue biomanufacturing.
Collapse
Affiliation(s)
- Zhanping Ren
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Anicca D Harriot
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA
| | - Devin B Mair
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA
| | | | - Peter H U Lee
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, 02912, USA
- Department of Cardiothoracic Surgery, Southcoast Health, Fall River, MA, 02720, USA
| | - Deok-Ho Kim
- Department of Mechanical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21205, USA
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Center for Microphysiological Systems, Johns Hopkins University, Baltimore, MD, 21205, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, 21218, USA
| |
Collapse
|
7
|
Ray N, Park SJ, Jung H, Kim J, Korcsmaros T, Moon Y. Stress-responsive Gdf15 counteracts renointestinal toxicity via autophagic and microbiota reprogramming. Commun Biol 2023; 6:602. [PMID: 37270567 DOI: 10.1038/s42003-023-04965-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 05/22/2023] [Indexed: 06/05/2023] Open
Abstract
The integrated stress response (ISR) plays a pivotal role in the cellular stress response, primarily through global translational arrest and the upregulation of cellular adaptation-linked molecules. Growth differentiation factor 15 (Gdf15) is a potent stress-responsive biomarker of clinical inflammatory and metabolic distress in various types of diseases. Herein, we assess whether ISR-driven cellular stress contributes to pathophysiological outcomes by modulating Gdf15. Clinical transcriptome analysis demonstrates that PKR is positively associated with Gdf15 expression in patients with renal injury. Gdf15 expression is dependent on protein kinase R (PKR)-linked ISR during acute renointestinal distress in mice and genetic ablation of Gdf15 aggravates chemical-induced lesions in renal tissues and the gut barrier. An in-depth evaluation of the gut microbiota indicates that Gdf15 is associated with the abundance of mucin metabolism-linked bacteria and their enzymes. Moreover, stress-responsive Gdf15 facilitates mucin production and cellular survival via the reorganization of the autophagy regulatory network. Collectively, ISR-activated Gdf15 counteracts pathological processes via the protective reprogramming of the autophagic network and microbial community, thereby providing robust predictive biomarkers and interventions against renointestinal distress.
Collapse
Affiliation(s)
- Navin Ray
- Laboratory of Mucosal Exposome and Biomodulation, Department of Integrative Biomedical Sciences, Pusan National University, Yangsan, Korea
| | - Seung Jun Park
- Laboratory of Mucosal Exposome and Biomodulation, Department of Integrative Biomedical Sciences, Pusan National University, Yangsan, Korea
| | - Hoyung Jung
- Laboratory of Mucosal Exposome and Biomodulation, Department of Integrative Biomedical Sciences, Pusan National University, Yangsan, Korea
| | - Juil Kim
- Laboratory of Mucosal Exposome and Biomodulation, Department of Integrative Biomedical Sciences, Pusan National University, Yangsan, Korea
| | - Tamas Korcsmaros
- Division of Digestive Diseases, Faculty of Medicine, Imperial College London, London, UK
- Earlham Institute, Norwich Research Park, Norwich, UK
- Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Yuseok Moon
- Laboratory of Mucosal Exposome and Biomodulation, Department of Integrative Biomedical Sciences, Pusan National University, Yangsan, Korea.
- Earlham Institute, Norwich Research Park, Norwich, UK.
- Graduate Program of Genomic Data Sciences, Pusan National University, Yangsan, Korea.
| |
Collapse
|
8
|
Ramírez-Flores CJ, Tibabuzo Perdomo AM, Gallego-López GM, Knoll LJ. Transcending Dimensions in Apicomplexan Research: from Two-Dimensional to Three-Dimensional In Vitro Cultures. Microbiol Mol Biol Rev 2022; 86:e0002522. [PMID: 35412359 PMCID: PMC9199416 DOI: 10.1128/mmbr.00025-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Parasites belonging to the Apicomplexa phylum are among the most successful pathogens known in nature. They can infect a wide range of hosts, often remain undetected by the immune system, and cause acute and chronic illness. In this phylum, we can find parasites of human and veterinary health relevance, such as Toxoplasma, Plasmodium, Cryptosporidium, and Eimeria. There are still many unknowns about the biology of these pathogens due to the ethical and practical issues of performing research in their natural hosts. Animal models are often difficult or nonexistent, and as a result, there are apicomplexan life cycle stages that have not been studied. One recent alternative has been the use of three-dimensional (3D) systems such as organoids, 3D scaffolds with different matrices, microfluidic devices, organs-on-a-chip, and other tissue culture models. These 3D systems have facilitated and expanded the research of apicomplexans, allowing us to explore life stages that were previously out of reach and experimental procedures that were practically impossible to perform in animal models. Human- and animal-derived 3D systems can be obtained from different organs, allowing us to model host-pathogen interactions for diagnostic methods and vaccine development, drug testing, exploratory biology, and other applications. In this review, we summarize the most recent advances in the use of 3D systems applied to apicomplexans. We show the wide array of strategies that have been successfully used so far and apply them to explore other organisms that have been less studied.
Collapse
Affiliation(s)
- Carlos J. Ramírez-Flores
- Department of Medical Microbiology and Immunology, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Andrés M. Tibabuzo Perdomo
- Department of Medical Microbiology and Immunology, University of Wisconsin–Madison, Madison, Wisconsin, USA
| | - Gina M. Gallego-López
- Department of Medical Microbiology and Immunology, University of Wisconsin–Madison, Madison, Wisconsin, USA
- Morgridge Institute for Research, Madison, Wisconsin, USA
| | - Laura J. Knoll
- Department of Medical Microbiology and Immunology, University of Wisconsin–Madison, Madison, Wisconsin, USA
| |
Collapse
|
9
|
Barrila J, Yang J, Franco Meléndez KP, Yang S, Buss K, Davis TJ, Aronow BJ, Bean HD, Davis RR, Forsyth RJ, Ott CM, Gangaraju S, Kang BY, Hanratty B, Nydam SD, Nauman EA, Kong W, Steel J, Nickerson CA. Spaceflight Analogue Culture Enhances the Host-Pathogen Interaction Between Salmonella and a 3-D Biomimetic Intestinal Co-Culture Model. Front Cell Infect Microbiol 2022; 12:705647. [PMID: 35711662 PMCID: PMC9195300 DOI: 10.3389/fcimb.2022.705647] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 04/29/2022] [Indexed: 11/13/2022] Open
Abstract
Physical forces associated with spaceflight and spaceflight analogue culture regulate a wide range of physiological responses by both bacterial and mammalian cells that can impact infection. However, our mechanistic understanding of how these environments regulate host-pathogen interactions in humans is poorly understood. Using a spaceflight analogue low fluid shear culture system, we investigated the effect of Low Shear Modeled Microgravity (LSMMG) culture on the colonization of Salmonella Typhimurium in a 3-D biomimetic model of human colonic epithelium containing macrophages. RNA-seq profiling of stationary phase wild type and Δhfq mutant bacteria alone indicated that LSMMG culture induced global changes in gene expression in both strains and that the RNA binding protein Hfq played a significant role in regulating the transcriptional response of the pathogen to LSMMG culture. However, a core set of genes important for adhesion, invasion, and motility were commonly induced in both strains. LSMMG culture enhanced the colonization (adherence, invasion and intracellular survival) of Salmonella in this advanced model of intestinal epithelium using a mechanism that was independent of Hfq. Although S. Typhimurium Δhfq mutants are normally defective for invasion when grown as conventional shaking cultures, LSMMG conditions unexpectedly enabled high levels of colonization by an isogenic Δhfq mutant. In response to infection with either the wild type or mutant, host cells upregulated transcripts involved in inflammation, tissue remodeling, and wound healing during intracellular survival. Interestingly, infection by the Δhfq mutant led to fewer transcriptional differences between LSMMG- and control-infected host cells relative to infection with the wild type strain. This is the first study to investigate the effect of LSMMG culture on the interaction between S. Typhimurium and a 3-D model of human intestinal tissue. These findings advance our understanding of how physical forces can impact the early stages of human enteric salmonellosis.
Collapse
Affiliation(s)
- Jennifer Barrila
- Biodesign Center for Fundamental and Applied Microbiomics, Arizona State University, Tempe, AZ, United States
- *Correspondence: Jennifer Barrila, ; Cheryl A. Nickerson,
| | - Jiseon Yang
- Biodesign Center for Fundamental and Applied Microbiomics, Arizona State University, Tempe, AZ, United States
| | - Karla P. Franco Meléndez
- Biodesign Center for Fundamental and Applied Microbiomics, Arizona State University, Tempe, AZ, United States
- School of Life Sciences, Arizona State University, Tempe, AZ, United States
- Genomics and Bioinformatics Research Unit, Agricultural Research Service, U.S. Department of Agriculture, Gainesville, FL, United States
| | - Shanshan Yang
- Bioinformatics Core Facility, Bioscience, Knowledge Enterprise, Arizona State University, Tempe, AZ, United States
| | - Kristina Buss
- Bioinformatics Core Facility, Bioscience, Knowledge Enterprise, Arizona State University, Tempe, AZ, United States
| | - Trenton J. Davis
- Biodesign Center for Fundamental and Applied Microbiomics, Arizona State University, Tempe, AZ, United States
- School of Life Sciences, Arizona State University, Tempe, AZ, United States
| | - Bruce J. Aronow
- Division of Biomedical Informatics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, United States
| | - Heather D. Bean
- Biodesign Center for Fundamental and Applied Microbiomics, Arizona State University, Tempe, AZ, United States
- School of Life Sciences, Arizona State University, Tempe, AZ, United States
| | - Richard R. Davis
- Biodesign Center for Fundamental and Applied Microbiomics, Arizona State University, Tempe, AZ, United States
| | - Rebecca J. Forsyth
- Biodesign Center for Fundamental and Applied Microbiomics, Arizona State University, Tempe, AZ, United States
| | - C. Mark Ott
- Biomedical Research and Environmental Sciences Division, NASA Johnson Space Center, Houston, TX, United States
| | - Sandhya Gangaraju
- Biodesign Center for Fundamental and Applied Microbiomics, Arizona State University, Tempe, AZ, United States
| | - Bianca Y. Kang
- Biodesign Center for Fundamental and Applied Microbiomics, Arizona State University, Tempe, AZ, United States
| | - Brian Hanratty
- Bioinformatics Core Facility, Bioscience, Knowledge Enterprise, Arizona State University, Tempe, AZ, United States
| | - Seth D. Nydam
- Department of Animal Care & Technologies, Arizona State University, Tempe, AZ, United States
| | - Eric A. Nauman
- School of Mechanical Engineering, Weldon School of Biomedical Engineering and Department of Basic Medical Sciences, Purdue University, West Lafayette, IN, United States
| | - Wei Kong
- Biodesign Center for Immunotherapy, Vaccines and Virotherapy, Arizona State University, Tempe, AZ, United States
| | - Jason Steel
- Bioinformatics Core Facility, Bioscience, Knowledge Enterprise, Arizona State University, Tempe, AZ, United States
| | - Cheryl A. Nickerson
- Biodesign Center for Fundamental and Applied Microbiomics, Arizona State University, Tempe, AZ, United States
- School of Life Sciences, Arizona State University, Tempe, AZ, United States
- *Correspondence: Jennifer Barrila, ; Cheryl A. Nickerson,
| |
Collapse
|
10
|
The Oesophageal Squamous Cell Carcinoma Cell Line COLO-680N Fails to Support Sustained Cryptosporidium parvum Proliferation. Pathogens 2021; 11:pathogens11010049. [PMID: 35055997 PMCID: PMC8778297 DOI: 10.3390/pathogens11010049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/28/2021] [Accepted: 12/30/2021] [Indexed: 11/16/2022] Open
Abstract
Cryptosporidium parvum is an important diarrhoea-associated protozoan, which is difficult to propagate in vitro. In 2017, a report described a continuous culture of C. parvum Moredun strain, in the oesophageal squamous cell carcinoma cell line COLO-680N, as an easy-to-use system for C. parvum propagation and continuous production of oocysts. Here, we report that—using the Köllitsch strain of C. parvum—even though COLO-680N cells, indeed, allowed parasite invasion and early asexual parasite replication, C. parvum proliferation decreased after the second day post infection. Considering recurring studies, reporting on successful production of newly generated Cryptosporidium oocysts in the past, and the subsequent replication failure by other research groups, the current data stand as a reminder of the importance of reproducibility of in vitro systems in cryptosporidiosis research. This is of special importance since it will only be possible to develop promising strategies to fight cryptosporidiosis and its ominous consequences for both human and animal health by a continuous and reliable methodological progress.
Collapse
|
11
|
First Metabolic Insights into Ex Vivo Cryptosporidium parvum-Infected Bovine Small Intestinal Explants Studied under Physioxic Conditions. BIOLOGY 2021; 10:biology10100963. [PMID: 34681062 PMCID: PMC8533177 DOI: 10.3390/biology10100963] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 09/14/2021] [Accepted: 09/22/2021] [Indexed: 12/12/2022]
Abstract
Simple Summary As the most relevant zoonotic cause of cryptosporidiosis, C. parvum infects cattle worldwide. In vitro studies on C. parvum are absent on the most important animal host under physiological oxygen conditions of the intestine. The aim of this study was to rectify this lack of knowledge, and to deliver a practical model to study C. parvum–host cell–intestinal microbiome interactions in the metabolic context. The present metabolic analyses of C. parvum-infected bovine small intestinal (BSI)-explants revealed a parasite-dependent reduction in important metabolic activities (e.g., glycolysis, glutaminolysis) at 3 hpi (hours post-infection) followed by striking increases in the same metabolic functions at 6 hpi, thus paralleling previously reported metabolic impacts of C. parvum on humans. In addition, PCA analysis confirmed physiological oxygen concentrations as a driving factor of metabolic responses in infected BSI explants. The present model allows the study of C. parvum-triggered metabolic modulation of intestinal cells. Moreover, this realistic platform offers the possibility to address pending questions regarding C. parvum–host cell–intestinal microbiome interactions. Thus, the present approach may deliver important insights into how to promote the innate immune system–intestinal microbiome alliances, which maintain the epithelial integrity of the gut thereby supporting human and animal health. Abstract The apicomplexan Cryptosporidium parvum causes thousands of human deaths yearly. Since bovines represent the most important reservoir of C. parvum, the analysis of infected bovine small intestinal (BSI) explants cultured under physioxia offers a realistic model to study C. parvum–host cell–microbiome interactions. Here, C. parvum-infected BSI explants and primary bovine small intestinal epithelial cells were analysed for parasite development and metabolic reactions. Metabolic conversion rates in supernatants of BSI explants were measured after infection, documenting an immediate parasite-driven metabolic interference. Given that oxygen concentrations affect cellular metabolism, measurements were performed at both 5% O2 (physiological intestinal conditions) and 21% O2 (commonly used, hyperoxic lab conditions). Overall, analyses of C. parvum-infected BSI explants revealed a downregulation of conversion rates of key metabolites—such as glucose, lactate, pyruvate, alanine, and aspartate—at 3 hpi, followed by a rapid increase in the same conversion rates at 6 hpi. Moreover, PCA revealed physioxia as a driving factor of metabolic responses in C. parvum-infected BSI explants. Overall, the ex vivo model described here may allow scientists to address pending questions as to how host cell–microbiome alliances influence intestinal epithelial integrity and support the development of protective intestinal immune reactions against C. parvum infections in a realistic scenario under physioxic conditions.
Collapse
|
12
|
O'Leary JK, Sleator RD, Lucey B. Cryptosporidium spp. diagnosis and research in the 21 st century. Food Waterborne Parasitol 2021; 24:e00131. [PMID: 34471706 PMCID: PMC8390533 DOI: 10.1016/j.fawpar.2021.e00131] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 08/06/2021] [Accepted: 08/17/2021] [Indexed: 01/01/2023] Open
Abstract
The protozoan parasite Cryptosporidium has emerged as a leading cause of diarrhoeal illness worldwide, posing a significant threat to young children and immunocompromised patients. While endemic in the vast majority of developing countries, Cryptosporidium also has the potential to cause waterborne epidemics and large scale outbreaks in both developing and developed nations. Anthroponontic and zoonotic transmission routes are well defined, with the ingestion of faecally contaminated food and water supplies a common source of infection. Microscopy, the current diagnostic mainstay, is considered by many to be suboptimal. This has prompted a shift towards alternative diagnostic techniques in the advent of the molecular era. Molecular methods, particularly PCR, are gaining traction in a diagnostic capacity over microscopy in the diagnosis of cryptosporidiosis, given the laborious and often tedious nature of the latter. Until now, developments in the field of Cryptosporidium detection and research have been somewhat hampered by the intractable nature of this parasite. However, recent advances in the field have taken the tentative first steps towards bringing Cryptosporidium research into the 21st century. Herein, we provide a review of these advances.
Collapse
Affiliation(s)
- Jennifer K. O'Leary
- Department of Biological Sciences, Munster Technological University, Bishopstown Campus, Cork, Ireland
| | - Roy D. Sleator
- Department of Biological Sciences, Munster Technological University, Bishopstown Campus, Cork, Ireland
| | - Brigid Lucey
- Department of Biological Sciences, Munster Technological University, Bishopstown Campus, Cork, Ireland
| |
Collapse
|
13
|
Crawford CK, Kol A. The Mucosal Innate Immune Response to Cryptosporidium parvum, a Global One Health Issue. Front Cell Infect Microbiol 2021; 11:689401. [PMID: 34113580 PMCID: PMC8185216 DOI: 10.3389/fcimb.2021.689401] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 05/07/2021] [Indexed: 02/02/2023] Open
Abstract
Cryptosporidium parvum is an apicomplexan parasite that infects the intestinal epithelium of humans and livestock animals worldwide. Cryptosporidiosis is a leading cause of diarrheal-related deaths in young children and a major cause of economic loss in cattle operations. The disease is especially dangerous to infants and immunocompromised individuals, for which there is no effective treatment or vaccination. As human-to-human, animal-to-animal and animal-to-human transmission play a role in cryptosporidiosis disease ecology, a holistic 'One Health' approach is required for disease control. Upon infection, the host's innate immune response restricts parasite growth and initiates the adaptive immune response, which is necessary for parasite clearance and recovery. The innate immune response involves a complex communicative interplay between epithelial and specialized innate immune cells. Traditional models have been used to study innate immune responses to C. parvum but cannot fully recapitulate natural host-pathogen interactions. Recent shifts to human and bovine organoid cultures are enabling deeper understanding of host-specific innate immunity response to infection. This review examines recent advances and highlights research gaps in our understanding of the host-specific innate immune response to C. parvum. Furthermore, we discuss evolving research models used in the field and potential developments on the horizon.
Collapse
Affiliation(s)
- Charles K Crawford
- Department of Pathology, Microbiology, & Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Amir Kol
- Department of Pathology, Microbiology, & Immunology, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| |
Collapse
|
14
|
Pance A. The Stem Cell Revolution Revealing Protozoan Parasites' Secrets and Paving the Way towards Vaccine Development. Vaccines (Basel) 2021; 9:105. [PMID: 33572549 PMCID: PMC7911700 DOI: 10.3390/vaccines9020105] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/26/2021] [Accepted: 01/27/2021] [Indexed: 12/13/2022] Open
Abstract
Protozoan infections are leading causes of morbidity and mortality in humans and some of the most important neglected diseases in the world. Despite relentless efforts devoted to vaccine and drug development, adequate tools to treat and prevent most of these diseases are still lacking. One of the greatest hurdles is the lack of understanding of host-parasite interactions. This gap in our knowledge comes from the fact that these parasites have complex life cycles, during which they infect a variety of specific cell types that are difficult to access or model in vitro. Even in those cases when host cells are readily available, these are generally terminally differentiated and difficult or impossible to manipulate genetically, which prevents assessing the role of human factors in these diseases. The advent of stem cell technology has opened exciting new possibilities to advance our knowledge in this field. The capacity to culture Embryonic Stem Cells, derive Induced Pluripotent Stem Cells from people and the development of protocols for differentiation into an ever-increasing variety of cell types and organoids, together with advances in genome editing, represent a huge resource to finally crack the mysteries protozoan parasites hold and unveil novel targets for prevention and treatment.
Collapse
Affiliation(s)
- Alena Pance
- The Wellcome Sanger Institute, Genome Campus, Hinxton Cambridgeshire CB10 1SA, UK
| |
Collapse
|
15
|
Advanced 3D Cell Culture Techniques in Micro-Bioreactors, Part II: Systems and Applications. Processes (Basel) 2020. [DOI: 10.3390/pr9010021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
In this second part of our systematic review on the research area of 3D cell culture in micro-bioreactors we give a detailed description of the published work with regard to the existing micro-bioreactor types and their applications, and highlight important results gathered with the respective systems. As an interesting detail, we found that micro-bioreactors have already been used in SARS-CoV research prior to the SARS-CoV2 pandemic. As our literature research revealed a variety of 3D cell culture configurations in the examined bioreactor systems, we defined in review part one “complexity levels” by means of the corresponding 3D cell culture techniques applied in the systems. The definition of the complexity is thereby based on the knowledge that the spatial distribution of cell-extracellular matrix interactions and the spatial distribution of homologous and heterologous cell–cell contacts play an important role in modulating cell functions. Because at least one of these parameters can be assigned to the 3D cell culture techniques discussed in the present review, we structured the studies according to the complexity levels applied in the MBR systems.
Collapse
|
16
|
Advanced 3D Cell Culture Techniques in Micro-Bioreactors, Part I: A Systematic Analysis of the Literature Published between 2000 and 2020. Processes (Basel) 2020. [DOI: 10.3390/pr8121656] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Bioreactors have proven useful for a vast amount of applications. Besides classical large-scale bioreactors and fermenters for prokaryotic and eukaryotic organisms, micro-bioreactors, as specialized bioreactor systems, have become an invaluable tool for mammalian 3D cell cultures. In this systematic review we analyze the literature in the field of eukaryotic 3D cell culture in micro-bioreactors within the last 20 years. For this, we define complexity levels with regard to the cellular 3D microenvironment concerning cell–matrix-contact, cell–cell-contact and the number of different cell types present at the same time. Moreover, we examine the data with regard to the micro-bioreactor design including mode of cell stimulation/nutrient supply and materials used for the micro-bioreactors, the corresponding 3D cell culture techniques and the related cellular microenvironment, the cell types and in vitro models used. As a data source we used the National Library of Medicine and analyzed the studies published from 2000 to 2020.
Collapse
|
17
|
Bhalchandra S, Lamisere H, Ward H. Intestinal organoid/enteroid-based models for Cryptosporidium. Curr Opin Microbiol 2020; 58:124-129. [PMID: 33113480 PMCID: PMC7758878 DOI: 10.1016/j.mib.2020.10.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 09/16/2020] [Accepted: 10/02/2020] [Indexed: 02/08/2023]
Abstract
Cryptosporidium is a leading cause of diarrhea and death in young children and untreated AIDS patients in resource-poor settings, and of waterborne outbreaks of disease in developed countries. However, there is no consistently effective treatment for vulnerable populations. Progress towards development of therapeutics for cryptosporidiosis has been hampered by lack of optimal culture systems to study it. New advances in organoid/enteroid technology have contributed to improved platforms to culture and propagate Cryptosporidium. Here we discuss recent breakthroughs in the field and highlight different models for functional ex vivo organoid or enteroidderived culture systems. These systems will lead to a better understanding of the mechanisms of host-parasite interactions in vivo.
Collapse
Affiliation(s)
- Seema Bhalchandra
- Tufts Medical Center, Tufts University School of Medicine, Boston, MA, USA.
| | - Hymlaire Lamisere
- Tufts University Graduate School of Biomedical Sciences, Boston, MA, USA
| | - Honorine Ward
- Tufts Medical Center, Tufts University School of Medicine, Boston, MA, USA; Tufts University Graduate School of Biomedical Sciences, Boston, MA, USA
| |
Collapse
|
18
|
Park SH, Moon Y. Enterocyte-Based Bioassay via Quantitative Combination of Proinflammatory Sentinels Specific to 8-keto-trichothecenes. Front Immunol 2020; 11:1530. [PMID: 32765531 PMCID: PMC7378738 DOI: 10.3389/fimmu.2020.01530] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 06/10/2020] [Indexed: 12/12/2022] Open
Abstract
Type B 8-keto-trichothecenes are muco-active mycotoxins that exist as inevitable contaminants in cereal-based foodstuffs. Gut-associated inflammation is an early frontline response during human and animal exposure to these mycotoxins. Despite various tools for chemical identification, optimized biomonitoring of sentinel response-associated biomarkers is required to assess the specific proinflammatory actions of 8-keto-trichothecenes in the gut epithelial barrier. In the present study, intoxication with 8-keto-trichothecenes in human intestinal epithelial cells was found to trigger early response gene 1 product (EGR-1) that plays crucial roles in proinflammatory chemokine induction. In contrast, epithelial exposure to 8-keto-trichothecenes resulted in downregulated expression of nuclear factor NF-kappa-B p65 protein, a key transcription factor, during general inflammatory responses in the gut. Based on the early molecular patterns of expression, the inflammation-inducing activity of 8-keto-trichothecenes was quantified using intestinal epithelial cells with dual reporters for EGR-1 and p65 proteins. EGR-1-responsive elements were linked to luciferase reporter while p65 promoter was bound to secretory alkaline phosphatase (SEAP) reporter. In response to conventional inflammagens such as endotoxins and cytokines such as TNF-α, both luciferase and SEAP activity were elevated in a dose-dependent manner. However, as expected from the mechanistic evaluation, 8-keto-trichothecene-exposed dual reporters of luciferase and SEAP displayed contrasting expression patterns. Furthermore, 8-keto-trichothecene-elevated EGR-1-responsive luciferase activity was improved by deficiency of PSMA3, an α-type subunit of the 20S proteasome core complex for ubiquitin-dependent EGR-1 degradation. This molecular event-based dual biomonitoring in epithelial cells is a promising supplementary tool for detecting typical molecular inflammatory pathways in response to 8-keto-trichothecenes in the food matrix.
Collapse
Affiliation(s)
- Seong-Hwan Park
- Laboratory of Mucosal Exposome and Biomodulation, Department of Biomedical Sciences, Pusan National University, Yangsan, South Korea
| | - Yuseok Moon
- Laboratory of Mucosal Exposome and Biomodulation, Department of Biomedical Sciences, Pusan National University, Yangsan, South Korea.,Biomedical Research Institute, Pusan National University, Yangsan, South Korea
| |
Collapse
|
19
|
Caveolar communication with xenobiotic-stalled ribosomes compromises gut barrier integrity. Commun Biol 2020; 3:270. [PMID: 32461676 PMCID: PMC7253476 DOI: 10.1038/s42003-020-0994-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 05/04/2020] [Indexed: 12/15/2022] Open
Abstract
In response to internal and external insults, the intestinal lining undergoes various types of epithelial adaptation or pathologic distress via stress-responsive eIF2α kinase signaling and subsequent cellular reprogramming. As a vital platform for growth factor-linked adaptive signaling, caveolae were evaluated for epithelial modulation of the insulted gut. Patients under ulcerative insult displayed enhanced expression of caveolin-1, the main structural component of caveolae, which was positively associated with expression of protein kinase R (PKR), the ribosomal stress-responsive eIF2α kinase. PKR-linked biological responses were simulated in experimental gut models of ribosome-inactivating stress using mice and Caenorhabditis elegans. Caveolar activation counteracted the expression of wound-protective epidermal growth factor receptor (EGFR) and its target genes, such as chemokines that were pivotal for epithelial integrity in the ribosome-inactivated gut. Mechanistic findings regarding ribosomal inactivation-associated disorders in the gut barrier provide crucial molecular evidence for detrimental caveolar actions against EGFR-mediated epithelial protection in patients with IBD.
Collapse
|
20
|
Gunasekera S, Zahedi A, O’Dea M, King B, Monis P, Thierry B, M. Carr J, Ryan U. Organoids and Bioengineered Intestinal Models: Potential Solutions to the Cryptosporidium Culturing Dilemma. Microorganisms 2020; 8:microorganisms8050715. [PMID: 32403447 PMCID: PMC7285185 DOI: 10.3390/microorganisms8050715] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 05/06/2020] [Accepted: 05/08/2020] [Indexed: 12/19/2022] Open
Abstract
Cryptosporidium is a major cause of severe diarrhea-related disease in children in developing countries, but currently no vaccine or effective treatment exists for those who are most at risk of serious illness. This is partly due to the lack of in vitro culturing methods that are able to support the entire Cryptosporidium life cycle, which has led to research in Cryptosporidium biology lagging behind other protozoan parasites. In vivo models such as gnotobiotic piglets are complex, and standard in vitro culturing methods in transformed cell lines, such as HCT-8 cells, have not been able to fully support fertilization occurring in vitro. Additionally, the Cryptosporidium life cycle has also been reported to occur in the absence of host cells. Recently developed bioengineered intestinal models, however, have shown more promising results and are able to reproduce a whole cycle of infectivity in one model system. This review evaluates the recent advances in Cryptosporidium culturing techniques and proposes future directions for research that may build upon these successes.
Collapse
Affiliation(s)
- Samantha Gunasekera
- Vector and Waterborne Pathogens Research Group, College of Science, Health, Engineering and Education, Murdoch University, Murdoch 6150, Western Australia, Australia;
- Correspondence: (S.G.); (U.R.); Tel.: +61-8-9360-2495 (S.G.); +61-8-9360-2482 (U.R.)
| | - Alireza Zahedi
- Vector and Waterborne Pathogens Research Group, College of Science, Health, Engineering and Education, Murdoch University, Murdoch 6150, Western Australia, Australia;
| | - Mark O’Dea
- Antimicrobial Resistance and Infectious Diseases Laboratory, College of Science, Health, Engineering and Education, Murdoch University, Murdoch 6150, Western Australia, Australia; m.o’
| | - Brendon King
- South Australian Water Corporation, Adelaide 5000, South Australia, Australia; (B.K.); (P.M.)
- College of Medicine and Public Health, Flinders University, Adelaide 5042, South Australia, Australia;
| | - Paul Monis
- South Australian Water Corporation, Adelaide 5000, South Australia, Australia; (B.K.); (P.M.)
- Future Industries Institute and ARC Centre of Excellence for Convergent Bio and Nano Science, University of South Australia, Adelaide 5095, South Australia, Australia;
| | - Benjamin Thierry
- Future Industries Institute and ARC Centre of Excellence for Convergent Bio and Nano Science, University of South Australia, Adelaide 5095, South Australia, Australia;
| | - Jillian M. Carr
- College of Medicine and Public Health, Flinders University, Adelaide 5042, South Australia, Australia;
| | - Una Ryan
- Vector and Waterborne Pathogens Research Group, College of Science, Health, Engineering and Education, Murdoch University, Murdoch 6150, Western Australia, Australia;
- Correspondence: (S.G.); (U.R.); Tel.: +61-8-9360-2495 (S.G.); +61-8-9360-2482 (U.R.)
| |
Collapse
|
21
|
Saleh F, Harb A, Soudani N, Zaraket H. A three-dimensional A549 cell culture model to study respiratory syncytial virus infections. J Infect Public Health 2020; 13:1142-1147. [PMID: 32360024 PMCID: PMC7190299 DOI: 10.1016/j.jiph.2020.03.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 03/10/2020] [Accepted: 03/16/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Respiratory syncytial virus (RSV) is a primary cause of morbidity and mortality worldwide, affecting infants, young children, and immune-compromised patients; however, currently no vaccine is available for prevention of RSV infections. The overwhelming majority of our knowledge of how RSV causes infection is based upon studies that have been carried out using traditional 2D methods, with cells cultured on flat plastic dishes. Although these simplified culture systems are essential to gain an insight into the fundamentals of host-pathogen interactions, cells in 2D are not exposed to the same conditions as cells in 3D tissues in the body and are therefore a poor representation of thein vivo microenvironment. In this study, we aim to develop the first 3D culture model for RSV infection using A549 cells to test its utility for RSV pathogenesis. METHODS To generate spheroids, A549 cells were cultured using ultra-low attachment plates to generate 25 × 103 cell spheroids. The viability of the spheroids was assessed by trypan blue exclusion assay and flow cytometry showing prominent live cells throughout the spheroids confirming high viability over seven days of incubation. RESULTS Immunostaining of A549 spheroids inoculated with RSV, showed time-dependent dissemination of the viral antigen RSV-F within the spheroid, resulting in syncytia formation and a 3-fold increase in mucin secretion compared to the uninfected cells. Additionally, RSV successfully replicated in the spheroids producing infectious virus as early as day one post-inoculation and was sustained for up to 7 days post-inoculation. CONCLUSIONS Results show that A549 spheroids are susceptible and permissive for RSV since they exhibit the characteristics of RSV infection including syncytia formation and mucin overexpression, suggesting that A549 spheroids can be used a promising model for studying RSV in vitro.
Collapse
Affiliation(s)
- Fatima Saleh
- Department of Medical Laboratory Sciences, Faculty of Health Sciences, Beirut Arab University, Lebanon
| | - Aya Harb
- Faculty of Medicine, Department of Experimental Pathology, Immunology & Microbiology, American University of Beirut, Beirut, Lebanon
| | - Nadia Soudani
- Faculty of Medicine, Department of Experimental Pathology, Immunology & Microbiology, American University of Beirut, Beirut, Lebanon; Faculty of Medicine, Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon; Doctoral School of Sciences and Technology, Research Platform for Environmental Science (PRASE), Faculty of Sciences, Lebanese University, Lebanon
| | - Hassan Zaraket
- Faculty of Medicine, Department of Experimental Pathology, Immunology & Microbiology, American University of Beirut, Beirut, Lebanon; Faculty of Medicine, Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon.
| |
Collapse
|
22
|
Kim KH, Lee SJ, Kim J, Moon Y. Dynamic Malignant Wave of Ribosome-Insulted Gut Niche via the Wnt-CTGF/CCN2 Circuit. iScience 2020; 23:101076. [PMID: 32361596 PMCID: PMC7200318 DOI: 10.1016/j.isci.2020.101076] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 12/13/2019] [Accepted: 04/14/2020] [Indexed: 12/21/2022] Open
Abstract
Stress-driven ribosome dysfunction triggers an eIF2α-mediated integrated stress response to maintain cellular homeostasis. Among four key eIF2α kinases, protein kinase R (PKR) expression positively associates with poor prognoses for colorectal cancer (CRC) patients. We identified PKR-linked Wnt signaling networks that facilitate early inflammatory niche and epithelial-mesenchymal transitions of tumor tissues in response to ribosomal insults. However, the downstream Wnt signaling target fibrogenic connective tissue growth factor (CTGF/CCN2) regulates the nuclear translocation of β-catenin in a negative feedback manner. Moreover, dwindling expression of the Wnt/β-catenin pathway-regulator CTGF triggers noncanonical Wnt pathway-mediated exacerbation of intestinal cancer progression such as an increase in cancer stemness and acquisition of chemoresistance in the presence of ribosomal insults. The Wnt-CTGF-circuit-associated landscape of oncogenic signaling events was verified with clinical genomic profiling. This ribosome-associated wave of crosstalk between stress and oncogenes provides valuable insight into potential molecular interventions against intestinal malignancies. PKR expression positively associates with poor prognoses for CRC patients CTGF/CCN2 mediates tumor niche remodeling under PKR-activating ribosomal stress CTGF/CCN2 antagonism of Wnt regulates cancer stemness and chemoresistance
Collapse
Affiliation(s)
- Ki Hyung Kim
- Laboratory of Mucosal Exposome and Biomodulation, Department of Biomedical Sciences, Pusan National University, Yangsan 50612, Korea; Department of Obstetrics and Gynecology, Pusan National University College of Medicine, Busan 49241, Korea; Biomedical Research Institute, Pusan National University Hospital, Busan, 49241, Korea
| | - Seung Joon Lee
- Laboratory of Mucosal Exposome and Biomodulation, Department of Biomedical Sciences, Pusan National University, Yangsan 50612, Korea
| | - Juil Kim
- Laboratory of Mucosal Exposome and Biomodulation, Department of Biomedical Sciences, Pusan National University, Yangsan 50612, Korea
| | - Yuseok Moon
- Laboratory of Mucosal Exposome and Biomodulation, Department of Biomedical Sciences, Pusan National University, Yangsan 50612, Korea; Biomedical Research Institute, Pusan National University Hospital, Busan, 49241, Korea.
| |
Collapse
|
23
|
The transcriptome of Cryptosporidium oocysts and intracellular stages. Sci Rep 2019; 9:7856. [PMID: 31133645 PMCID: PMC6536522 DOI: 10.1038/s41598-019-44289-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 05/14/2019] [Indexed: 01/24/2023] Open
Abstract
Human cryptosporidiosis is caused primarily by two species of apicomplexan parasites, Cryptosporidium parvum and C. hominis. Although infection of cell monolayers with sporozoites does not support the complete parasite life cycle, the in vitro system is used to study the asexual phase of multiplication, which consists of two generations of merogony. To better understand host-parasite interaction and to gain insight into gene regulatory processes driving the complex life cycle of Cryptosporidium parasites, we analyzed the transcriptome of C. parvum in oocysts, sporozoites and infected cell monolayers 2–48 h post-infection. Analysis of RNA-Seq data from replicate oocyst, sporozoite and intracellular samples revealed significant differences between transcriptomes expressed outside and inside the host cell. Compared to the transcriptome found in the host cell, the oocyst transcriptome is less diverse. Biological processes significantly over-represented intracellularly relate to biosynthetic processes. Genes significantly overexpressed in oocysts show evidence of specialized functions not found in other Apicomplexa. A more comprehensive view of gene regulation during the Cryptosporidium life cycle will require the analysis of later time points during the infection, particularly of the poorly studied sexual phase of the life cycle.
Collapse
|
24
|
Bones AJ, Jossé L, More C, Miller CN, Michaelis M, Tsaousis AD. Past and future trends of Cryptosporidium in vitro research. Exp Parasitol 2018; 196:28-37. [PMID: 30521793 PMCID: PMC6333944 DOI: 10.1016/j.exppara.2018.12.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Revised: 11/09/2018] [Accepted: 12/02/2018] [Indexed: 12/15/2022]
Abstract
Cryptosporidium is a genus of single celled parasites capable of infecting a wide range of animals including humans. Cryptosporidium species are members of the phylum apicomplexa, which includes well-known genera such as Plasmodium and Toxoplasma. Cryptosporidium parasites cause a severe gastro-intestinal disease known as cryptosporidiosis. They are one of the most common causes of childhood diarrhoea worldwide, and infection can have prolonged detrimental effects on the development of children, but also can be life threatening to HIV/AIDS patients and transplant recipients. A variety of hosts can act as reservoirs, and Cryptosporidium can persist in the environment for prolonged times as oocysts. While there has been substantial interest in these parasites, there is very little progress in terms of treatment development and understanding the majority of the life cycle of this unusual organism. In this review, we will provide an overview on the existing knowledge of the biology of the parasite and the current progress in developing in vitro cultivation systems. We will then describe a synopsis of current and next generation approaches that could spearhead further research in combating the parasite.
Collapse
Affiliation(s)
- Alexander J Bones
- Laboratory of Molecular and Evolutionary Parasitology, RAPID Group, School of Biosciences, University of Kent, Canterbury, Kent, UK
| | - Lyne Jossé
- Laboratory of Molecular and Evolutionary Parasitology, RAPID Group, School of Biosciences, University of Kent, Canterbury, Kent, UK
| | - Charlotte More
- Laboratory of Molecular and Evolutionary Parasitology, RAPID Group, School of Biosciences, University of Kent, Canterbury, Kent, UK
| | - Christopher N Miller
- Laboratory of Molecular and Evolutionary Parasitology, RAPID Group, School of Biosciences, University of Kent, Canterbury, Kent, UK
| | | | - Anastasios D Tsaousis
- Laboratory of Molecular and Evolutionary Parasitology, RAPID Group, School of Biosciences, University of Kent, Canterbury, Kent, UK; School of Biosciences, University of Kent, Canterbury, Kent, UK.
| |
Collapse
|
25
|
Modeling Host-Pathogen Interactions in the Context of the Microenvironment: Three-Dimensional Cell Culture Comes of Age. Infect Immun 2018; 86:IAI.00282-18. [PMID: 30181350 DOI: 10.1128/iai.00282-18] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Tissues and organs provide the structural and biochemical landscapes upon which microbial pathogens and commensals function to regulate health and disease. While flat two-dimensional (2-D) monolayers composed of a single cell type have provided important insight into understanding host-pathogen interactions and infectious disease mechanisms, these reductionist models lack many essential features present in the native host microenvironment that are known to regulate infection, including three-dimensional (3-D) architecture, multicellular complexity, commensal microbiota, gas exchange and nutrient gradients, and physiologically relevant biomechanical forces (e.g., fluid shear, stretch, compression). A major challenge in tissue engineering for infectious disease research is recreating this dynamic 3-D microenvironment (biological, chemical, and physical/mechanical) to more accurately model the initiation and progression of host-pathogen interactions in the laboratory. Here we review selected 3-D models of human intestinal mucosa, which represent a major portal of entry for infectious pathogens and an important niche for commensal microbiota. We highlight seminal studies that have used these models to interrogate host-pathogen interactions and infectious disease mechanisms, and we present this literature in the appropriate historical context. Models discussed include 3-D organotypic cultures engineered in the rotating wall vessel (RWV) bioreactor, extracellular matrix (ECM)-embedded/organoid models, and organ-on-a-chip (OAC) models. Collectively, these technologies provide a more physiologically relevant and predictive framework for investigating infectious disease mechanisms and antimicrobial therapies at the intersection of the host, microbe, and their local microenvironments.
Collapse
|
26
|
Trujillo-de Santiago G, Lobo-Zegers MJ, Montes-Fonseca SL, Zhang YS, Alvarez MM. Gut-microbiota-on-a-chip: an enabling field for physiological research. MICROPHYSIOLOGICAL SYSTEMS 2018; 2:7. [PMID: 33954286 PMCID: PMC8096182 DOI: 10.21037/mps.2018.09.01] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Overwhelming scientific evidence today confirms that the gut microbiota is a central player in human health. Knowledge about interactions between human gut microbiota and human health has evolved rapidly in the last decade, based on experimental work involving analysis of human fecal samples or animal models (mainly rodents). A more detailed and cost-effective description of this interplay is now being enabled by the use of in vitro systems (i.e., gut-microbiota-on-chip systems) that recapitulate key aspects of the interaction between microbiota and human cells. Here, we review recent examples of the design and use of pioneering on-chip platforms for the study of the cross-talk between representative members of human microbiota and human microtissues. In these systems, the combined use of state-of-the-art microfluidics, biomaterials, cell culture techniques, classical microbiology, and a touch of genetic expression profiling have converged for the development of gut-on-chip platforms capable of recreating key features of the interplay between human microbiota and host human tissues. We foresee that the integration of novel microfabrication techniques and stem cell technologies will further accelerate the development of more complex and physiologically relevant microbiota-on-chip platforms. In turn, this will foster the faster acquisition of knowledge regarding human microbiota and will enable important advances in the understanding of how to control or prevent disease.
Collapse
Affiliation(s)
- Grissel Trujillo-de Santiago
- Centro de Biotecnología-FEMSA, Tecnológico de Monterrey
- Departamento de Mecatrónica e Ingeniería Eléctrica, Campus Monterrey, CP 64849, Monterrey, Nuevo León, México
| | - Matías José Lobo-Zegers
- Centro de Biotecnología-FEMSA, Tecnológico de Monterrey
- Departamento de Mecatrónica e Ingeniería Eléctrica, Campus Monterrey, CP 64849, Monterrey, Nuevo León, México
| | - Silvia Lorena Montes-Fonseca
- Departamento de Bioingeniería, Escuela de Ingeniería y Ciencias, Tecnológico de Monterrey, CP 31300, Chihuahua, México
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, MA, USA
| | | |
Collapse
|
27
|
Li L, Chen B, Yan H, Zhao Y, Lou Z, Li J, Fu B, Zhu X, McManus DP, Dai J, Jia W. Three-dimensional hepatocyte culture system for the study of Echinococcus multilocularis larval development. PLoS Negl Trop Dis 2018. [PMID: 29538424 PMCID: PMC5868855 DOI: 10.1371/journal.pntd.0006309] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Background Hepatocyte-based metacestode culture is an attractive method to study alveolar echinococcosis (AE), but it is limited by the relatively short lifespan of cultured hepatocytes in maintaining their normal function. Methodology/principal findings We describe a three-dimensional (3D) hepatic culture system developed from co-cultured hepatocytes and mesenchymal stem cells using a collagen scaffold to study the development of Echinococcus multilocularis larvae. This 3D culture system preserved the function of hepatocytes for a longer period of time than their monolayer counterparts, with albumin secretion, 7-ethoxyresorufin O-deethylation activity, urea synthesis, CYP3A4 and CYP2D6 activity being highly preserved for 21–28 days. The expression levels of hepatocyte-specific genes including CLDN-3, Bsep, AFP, G6P, A1AT, CYP3A4 and NR1I3 were significantly higher in the 3D cultured system compared with their monolayer counterparts after 14-days in culture. Additionally, in the presence of 3D cultured hepatocytes, 81.2% of E. multilocularis protoscoleces rapidly de-differentiated into infective vesicles within eight weeks. Transcriptomic analyses revealed 807 differentially expressed genes between cultured vesicles and protoscoleces, including 119 genes uniquely expressed in protoscoleces, and 242 genes uniquely expressed in vesicles. These differentially expressed genes were mainly involved in parasite growth relating to the G-protein coupled receptor activity pathway, substrate-specific transmembrane transporter activity, cell-cell adhesion process, and potentially with neuroactive ligand-receptor interaction. Conclusions/significance This culture system provides a valuable advance in prolonging hepatocyte functionality, a foundation for future in-depth analysis of the host-parasite interaction in AE, and a useful model to evaluate potential therapeutic strategies to treat AE. Alveolar echinococcosis (AE) is one of the world’s most dangerous zoonoses. Although there have been recent advances in some aspects of the molecular biology of E. multilocularis, larval development is far from understood. An in vitro hepatocyte based cultivation system for the metacestode stage of E. multilocularis has been developed to improve our understanding of AE. However, in two-dimensional conventional cultures, hepatocytes rapidly lose key phenotypic and functional characteristics after only approximately seven days. This hinders long-term in vitro studies of E. multilocularis larvae, which require several months for development. Thus, in this paper, a three-dimensional (3D) hepatic model was developed for simulating the organotropism of E. multilocularis toward the liver of its intermediate host. This 3D model can preserve the functions of hepatocytes and results in rapidly developed E. multilocularis larva. Genes uniquely expressed in protoscoleces and vesicles provided key information for the further study of AE. The 3D hepatic model provides a new foundation for E. multilocularis developmental studies and in-depth analysis of the host-parasite interaction in AE.
Collapse
Affiliation(s)
- Li Li
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, P. R. China
| | - Bing Chen
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, P. R. China
| | - Hongbin Yan
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, P. R. China
| | - Yannan Zhao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, P. R. China
| | - Zhongzi Lou
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, P. R. China
| | - Jianqiu Li
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, P. R. China
| | - Baoquan Fu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, P. R. China
| | - Xingquan Zhu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, P. R. China
| | - Donald P. McManus
- Molecular Parasitology Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Jianwu Dai
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, P. R. China
- * E-mail: (JD); (WJ)
| | - Wanzhong Jia
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, P. R. China
- * E-mail: (JD); (WJ)
| |
Collapse
|
28
|
Kim J, Park SH, Do KH, Kim D, Moon Y. Interference with mutagenic aflatoxin B1-induced checkpoints through antagonistic action of ochratoxin A in intestinal cancer cells: a molecular explanation on potential risk of crosstalk between carcinogens. Oncotarget 2018; 7:39627-39639. [PMID: 27119350 PMCID: PMC5129958 DOI: 10.18632/oncotarget.8914] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 04/05/2016] [Indexed: 12/16/2022] Open
Abstract
Foodborne aflatoxin B1 (AFB1) and ochratoxin A (OTA) cause genotoxic injury and subsequent tumor formation. As a biomarker of oncogenic stimulation by genotoxic mycotoxins, p53-triggered Mdm2 was assessed in intestinal cancer cells. AFB1 increased Mdm2 reporter expression in a dose-dependent manner. However, this was strongly antagonized by OTA treatment. As a positive transcription factor of Mdm2 expression, p53 levels were also increased by AFB1 alone and reduced by OTA. With marginal cell death responses, AFB1 induced p53-mediated S phase arrest and cell cycle-regulating target genes, which was completely suppressed by OTA. Although enterocyte-dominant CYP3A5 counteracted AFB1-induced DNA damage, expression of CYP3A5 was decreased by OTA or AFB1. Instead, OTA enhanced expression of another metabolic inactivating enzyme CYP3A4, attenuation of formation of AFB1-DNA adduct and p53-mediated cell cycle checking responses to the mutagens. Finally, the growth of intestinal cancer cells exposed to the mycotoxin mixture significantly exceeded the expected growth calculated from that of cells treated with each mycotoxin. Although AFB1-induced mutagen formation was decreased by OTA, interference with checkpoints through antagonistic action of OTA may contribute to the survival of tumor cells with deleterious mutations by genotoxic mycotoxins, potently increasing the risk of carcinogenesis.
Collapse
Affiliation(s)
- Juil Kim
- Laboratory of Mucosal Exposome and Biomodulation, Department of Biomedical Sciences and Medical Research Institute, Pusan National University School of Medicine, Yangsan, South Korea
| | - Seong-Hwan Park
- Laboratory of Mucosal Exposome and Biomodulation, Department of Biomedical Sciences and Medical Research Institute, Pusan National University School of Medicine, Yangsan, South Korea
| | - Kee Hun Do
- Laboratory of Mucosal Exposome and Biomodulation, Department of Biomedical Sciences and Medical Research Institute, Pusan National University School of Medicine, Yangsan, South Korea
| | - Dongwook Kim
- National Institute of Animal Science, RDA, Wanju, South Korea
| | - Yuseok Moon
- Laboratory of Mucosal Exposome and Biomodulation, Department of Biomedical Sciences and Medical Research Institute, Pusan National University School of Medicine, Yangsan, South Korea.,Research Institute for Basic Sciences and Immunoregulatory Therapeutics Group in Brain Busan 21 Project, Pusan, South Korea
| |
Collapse
|
29
|
Melicherová J, Hofmannová L, Valigurová A. Response of cell lines to actual and simulated inoculation with Cryptosporidium proliferans. Eur J Protistol 2017; 62:101-121. [PMID: 29316479 DOI: 10.1016/j.ejop.2017.12.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 12/08/2017] [Accepted: 12/13/2017] [Indexed: 10/18/2022]
Abstract
The need for an effective treatment against cryptosporidiosis has triggered studies in the search for a working in vitro model. The peculiar niche of cryptosporidia at the brush border of host epithelial cells has been the subject of extensive debates. Despite extensive research on the invasion process, it remains enigmatic whether cryptosporidian host-parasite interactions result from an active invasion process or through encapsulation. We used HCT-8 and HT-29 cell lines for in vitro cultivation of the gastric parasite Cryptosporidium proliferans strain TS03. Using electron and confocal laser scanning microscopy, observations were carried out 24, 48 and 72 h after inoculation with a mixture of C. proliferans oocysts and sporozoites. Free sporozoites and putative merozoites were observed apparently searching for an appropriate infection site. Advanced stages, corresponding to trophozoites and meronts/gamonts enveloped by parasitophorous sac, and emptied sacs were detected. As our observations showed that even unexcysted oocysts became enveloped by cultured cell projections, using polystyrene microspheres, we evaluated the response of cell lines to simulated inoculation with cryptosporidian oocysts to verify innate and parasite-induced behaviour. We found that cultured cell encapsulation of oocysts is induced by parasite antigens, independent of any active invasion/motility.
Collapse
Affiliation(s)
- Janka Melicherová
- Department of Botany and Zoology, Faculty of Science, Masaryk University, Kotlářská 2, 611 37 Brno, Czech Republic
| | - Lada Hofmannová
- Department of Pathological Morphology and Parasitology, University of Veterinary and Pharmaceutical Sciences, Palackého tř. 1946/1, 612 42 Brno, Czech Republic
| | - Andrea Valigurová
- Department of Botany and Zoology, Faculty of Science, Masaryk University, Kotlářská 2, 611 37 Brno, Czech Republic.
| |
Collapse
|
30
|
Baydoun M, Vanneste SB, Creusy C, Guyot K, Gantois N, Chabe M, Delaire B, Mouray A, Baydoun A, Forzy G, Chieux V, Gosset P, Senez V, Viscogliosi E, Follet J, Certad G. Three-dimensional (3D) culture of adult murine colon as an in vitro model of cryptosporidiosis: Proof of concept. Sci Rep 2017; 7:17288. [PMID: 29230047 PMCID: PMC5725449 DOI: 10.1038/s41598-017-17304-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 11/19/2017] [Indexed: 01/12/2023] Open
Abstract
Cryptosporidium parvum is a major cause of diarrheal illness and was recently potentially associated with digestive carcinogenesis. Despite its impact on human health, Cryptosporidium pathogenesis remains poorly known, mainly due to the lack of a long-term culture method for this parasite. Thus, the aim of the present study was to develop a three-dimensional (3D) culture model from adult murine colon allowing biological investigations of the host-parasite interactions in an in vivo-like environment and, in particular, the development of parasite-induced neoplasia. Colonic explants were cultured and preserved ex vivo for 35 days and co-culturing was performed with C. parvum. Strikingly, the resulting system allowed the reproduction of neoplastic lesions in vitro at 27 days post-infection (PI), providing new evidence of the role of the parasite in the induction of carcinogenesis. This promising model could facilitate the study of host-pathogen interactions and the investigation of the process involved in Cryptosporidium-induced cell transformation.
Collapse
Affiliation(s)
- Martha Baydoun
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Lille, France.,ISA-YNCREA Hauts-de-France, Lille, France.,Univ. Lille, CNRS, ISEN, UMR 8520 - IEMN, Lille, France
| | - Sadia Benamrouz Vanneste
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Lille, France.,Laboratoire Ecologie et Biodiversité, Faculté de Gestion Economie et Sciences, Institut Catholique de Lille, Lille, France
| | - Colette Creusy
- Service d'Anatomie et de Cytologie Pathologiques, Groupement des Hopitaux de l'Institut Catholique de Lille (GHICL), Lille, France
| | - Karine Guyot
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Lille, France
| | - Nausicaa Gantois
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Lille, France
| | - Magali Chabe
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Lille, France.,Faculté de Pharmacie, Univ. de Lille, Lille, France
| | - Baptiste Delaire
- Service d'Anatomie et de Cytologie Pathologiques, Groupement des Hopitaux de l'Institut Catholique de Lille (GHICL), Lille, France
| | - Anthony Mouray
- Plateforme d'Expérimentations et de Hautes Technologies Animales, Institut Pasteur de Lille, Lille, France
| | - Atallah Baydoun
- Department of Internal Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA.,Department of Internal Medicine, Louis Stokes VA Medical Center, Cleveland, OH, USA.,Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Gerard Forzy
- Laboratoire de Biologie Médicale, Groupement des Hospitaux de l'Institut Catholique de Lille (GHICL), Lille, France
| | - Vincent Chieux
- Laboratoire de Biologie Médicale, Groupement des Hospitaux de l'Institut Catholique de Lille (GHICL), Lille, France
| | - Pierre Gosset
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Lille, France.,Service d'Anatomie et de Cytologie Pathologiques, Groupement des Hopitaux de l'Institut Catholique de Lille (GHICL), Lille, France
| | - Vincent Senez
- Univ. Lille, CNRS, ISEN, UMR 8520 - IEMN, Lille, France
| | - Eric Viscogliosi
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Lille, France
| | - Jérôme Follet
- ISA-YNCREA Hauts-de-France, Lille, France.,Univ. Lille, CNRS, ISEN, UMR 8520 - IEMN, Lille, France
| | - Gabriela Certad
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 8204 - CIIL - Centre d'Infection et d'Immunité de Lille, Lille, France. .,Département de la Recherche Médicale, Groupement des Hopitaux de l'Institut Catholique de Lille (GHICL), Faculté de Médecine et Maïeutique, Université Catholique de Lille, Lille, France.
| |
Collapse
|
31
|
Abstract
Cryptosporidium research has focused on the development of infection control, and effective therapy that has thus far been hampered by the inability to culture Cryptosporidium in vitro. Other limitations include inadequate animal models, cumbersome screening procedures for chemotherapeutic approaches and a lack of tools for genetic manipulation. These limitations can, however, be eased by the improvement and focused development of in vitro cultivation. The ability to culture relevant Cryptosporidium isolates in vitro and to propagate the life cycle stages that are responsible for causing disease in an infected host is still a critical link. This ability will facilitate other relevant approaches, e.g., the ability to knockout genes and the application of broader screening for drug discoveries and vaccine developments, in combination with new discoveries on the parasite's basic biology, genetic manipulation and new life cycle stages. Success in this effort represents an essential step towards significant progress in the control of cryptosporidiosis.
Collapse
|
32
|
Improvement of in vitro evaluation of chemical disinfectants for efficacy on Cryptosporidium parvum oocysts. Vet Parasitol 2017; 245:5-13. [PMID: 28969838 DOI: 10.1016/j.vetpar.2017.07.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 06/20/2017] [Accepted: 07/19/2017] [Indexed: 11/22/2022]
Abstract
Cryptosporidium parvum has been suggested as a suitable target for in vitro efficacy testing of disinfectants. To improve validity of a method based on exposure of HCT-8 monolayers to C. parvum oocysts we here critically evaluate and we propose certain procedural steps needed for the validation of disinfectants. Within a range of 0.02% to 0.4%, sodium taurocholate at 0.2% stimulated infection most efficiently while preserving host cell integrity. The course of invasion was monitored for periods of 30-240min post infection (p.i.). FACS analysis revealed that the proportion of sporozoites liberated from oocysts in the presence of 0.2% sodium taurocholate increased within 120min of incubation but remained constant thereafter. Maximum invasion of cells measured by qPCR was reached 180min p.i. and therefore set as invasion endpoint. As monolayers harvested 24h or 48h p.i. did not differ in the quantity of parasite hsp70 gene copies, DNA extraction can be performed as early as 24h p.i. Incubation of oocysts with 20% H2O2 for 2h resulted in inactivation of more than 99.5% both at room temperature and 10°C and appeared thus suitable as positive chemical treatment control. Four washing procedures considered to remove potentially toxic residual disinfectant from oocyst suspensions were tested. An application of a combination of DMSO (Dimethylsulfoxid), Tween20 and WSH (water of standardized hardness) appeared most efficient without deleterious effect of disinfectant residuals on the cell monolayer viability when oocysts accordingly washed were applied. In conclusion, for standardized in vitro evaluation of chemical disinfectants in C. parvum infected HTC-8 monolayers. (i) excystation medium should contain 0.2 % sodium taurocholate. (ii) excystation medium should be replaced by growth medium after 180 min. (iii) monolayers should be harvested 24 h p.i. for DNA preparation. (iv) ocysts exposed to 20 % H2O2 should be included as positive controls. (v) disinfected oocysts should be washed with DMSO/Tween20/WSH before they are transferred to monolayers.
Collapse
|
33
|
Barrila J, Yang J, Crabbé A, Sarker SF, Liu Y, Ott CM, Nelman-Gonzalez MA, Clemett SJ, Nydam SD, Forsyth RJ, Davis RR, Crucian BE, Quiriarte H, Roland KL, Brenneman K, Sams C, Loscher C, Nickerson CA. Three-dimensional organotypic co-culture model of intestinal epithelial cells and macrophages to study Salmonella enterica colonization patterns. NPJ Microgravity 2017; 3:10. [PMID: 28649632 PMCID: PMC5460263 DOI: 10.1038/s41526-017-0011-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 11/23/2016] [Accepted: 12/12/2016] [Indexed: 12/21/2022] Open
Abstract
Three-dimensional models of human intestinal epithelium mimic the differentiated form and function of parental tissues often not exhibited by two-dimensional monolayers and respond to Salmonella in key ways that reflect in vivo infections. To further enhance the physiological relevance of three-dimensional models to more closely approximate in vivo intestinal microenvironments encountered by Salmonella, we developed and validated a novel three-dimensional co-culture infection model of colonic epithelial cells and macrophages using the NASA Rotating Wall Vessel bioreactor. First, U937 cells were activated upon collagen-coated scaffolds. HT-29 epithelial cells were then added and the three-dimensional model was cultured in the bioreactor until optimal differentiation was reached, as assessed by immunohistochemical profiling and bead uptake assays. The new co-culture model exhibited in vivo-like structural and phenotypic characteristics, including three-dimensional architecture, apical-basolateral polarity, well-formed tight/adherens junctions, mucin, multiple epithelial cell types, and functional macrophages. Phagocytic activity of macrophages was confirmed by uptake of inert, bacteria-sized beads. Contribution of macrophages to infection was assessed by colonization studies of Salmonella pathovars with different host adaptations and disease phenotypes (Typhimurium ST19 strain SL1344 and ST313 strain D23580; Typhi Ty2). In addition, Salmonella were cultured aerobically or microaerobically, recapitulating environments encountered prior to and during intestinal infection, respectively. All Salmonella strains exhibited decreased colonization in co-culture (HT-29-U937) relative to epithelial (HT-29) models, indicating antimicrobial function of macrophages. Interestingly, D23580 exhibited enhanced replication/survival in both models following invasion. Pathovar-specific differences in colonization and intracellular co-localization patterns were observed. These findings emphasize the power of incorporating a series of related three-dimensional models within a study to identify microenvironmental factors important for regulating infection.
Collapse
Affiliation(s)
- Jennifer Barrila
- Center for Infectious Diseases and Vaccinology, The Biodesign Institute, Arizona State University, Tempe, AZ USA
| | - Jiseon Yang
- Center for Infectious Diseases and Vaccinology, The Biodesign Institute, Arizona State University, Tempe, AZ USA
| | - Aurélie Crabbé
- Center for Infectious Diseases and Vaccinology, The Biodesign Institute, Arizona State University, Tempe, AZ USA
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium
| | - Shameema F. Sarker
- Center for Infectious Diseases and Vaccinology, The Biodesign Institute, Arizona State University, Tempe, AZ USA
| | - Yulong Liu
- Center for Infectious Diseases and Vaccinology, The Biodesign Institute, Arizona State University, Tempe, AZ USA
| | - C. Mark Ott
- Biomedical Research and Environmental Sciences Division, NASA Johnson Space Center, Houston, TX USA
| | | | | | - Seth D. Nydam
- Center for Infectious Diseases and Vaccinology, The Biodesign Institute, Arizona State University, Tempe, AZ USA
| | - Rebecca J. Forsyth
- Center for Infectious Diseases and Vaccinology, The Biodesign Institute, Arizona State University, Tempe, AZ USA
| | - Richard R. Davis
- Center for Infectious Diseases and Vaccinology, The Biodesign Institute, Arizona State University, Tempe, AZ USA
| | - Brian E. Crucian
- Biomedical Research and Environmental Sciences Division, NASA Johnson Space Center, Houston, TX USA
| | | | - Kenneth L. Roland
- Center for Infectious Diseases and Vaccinology, The Biodesign Institute, Arizona State University, Tempe, AZ USA
| | - Karen Brenneman
- Center for Infectious Diseases and Vaccinology, The Biodesign Institute, Arizona State University, Tempe, AZ USA
| | - Clarence Sams
- Biomedical Research and Environmental Sciences Division, NASA Johnson Space Center, Houston, TX USA
| | - Christine Loscher
- Immunomodulation Research Group, School of Biotechnology, Dublin City University, Glasnevin, Ireland
| | - Cheryl A. Nickerson
- Center for Infectious Diseases and Vaccinology, The Biodesign Institute, Arizona State University, Tempe, AZ USA
- School of Life Sciences, Arizona State University, Tempe, AZ USA
| |
Collapse
|
34
|
Novel Bioengineered Three-Dimensional Human Intestinal Model for Long-Term Infection of Cryptosporidium parvum. Infect Immun 2017; 85:IAI.00731-16. [PMID: 28052996 DOI: 10.1128/iai.00731-16] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 12/23/2016] [Indexed: 01/08/2023] Open
Abstract
Cryptosporidium spp. are apicomplexan parasites of global importance that cause human diarrheal disease. In vitro culture models that may be used to study this parasite and that have physiological relevance to in vivo infection remain suboptimal. Thus, the pathogenesis of cryptosporidiosis remains poorly characterized, and interventions for the disease are limited. In this study, we evaluated the potential of a novel bioengineered three-dimensional (3D) human intestinal tissue model (which we developed previously) to support long-term infection by Cryptosporidium parvum Infection was assessed by immunofluorescence assays and confocal and scanning electron microscopy and quantified by quantitative reverse transcription-PCR. We found that C. parvum infected and developed in this tissue model for at least 17 days, the extent of the study time used in the present study. Contents from infected scaffolds could be transferred to fresh scaffolds to establish new infections for at least three rounds. Asexual and sexual stages and the formation of new oocysts were observed during the course of infection. Additionally, we observed ablation, blunting, or distortion of microvilli in infected epithelial cells. Ultimately, a 3D model system capable of supporting continuous Cryptosporidium infection will be a useful tool for the study of host-parasite interactions, identification of putative drug targets, screening of potential interventions, and propagation of genetically modified parasites.
Collapse
|
35
|
Park SH, Kim J, Yu M, Park JH, Kim YS, Moon Y. Epithelial Cholesterol Deficiency Attenuates Human Antigen R-linked Pro-inflammatory Stimulation via an SREBP2-linked Circuit. J Biol Chem 2016; 291:24641-24656. [PMID: 27703009 DOI: 10.1074/jbc.m116.723973] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 09/16/2016] [Indexed: 01/03/2023] Open
Abstract
Patients with chronic intestinal ulcerative diseases, such as inflammatory bowel disease, tend to exhibit abnormal lipid profiles, which may affect the gut epithelial integrity. We hypothesized that epithelial cholesterol depletion may trigger inflammation-checking machinery via cholesterol sentinel signaling molecules whose disruption in patients may aggravate inflammation and disease progression. In the present study, sterol regulatory element-binding protein 2 (SREBP2) as the cholesterol sentinel was assessed for its involvement in the epithelial inflammatory responses in cholesterol-depleted enterocytes. Patients and experimental animals with intestinal ulcerative injuries showed suppression in epithelial SREBP2. Moreover, SREBP2-deficient enterocytes showed enhanced pro-inflammatory signals in response to inflammatory insults, indicating regulatory roles of SREBP2 in gut epithelial inflammation. However, epithelial cholesterol depletion transiently induced pro-inflammatory chemokine expression regardless of the well known pro-inflammatory nuclear factor-κB signals. In contrast, cholesterol depletion also exerts regulatory actions to maintain epithelial homeostasis against excessive inflammation via SREBP2-associated signals in a negative feedback loop. Mechanistically, SREBP2 and its induced target EGR-1 were positively involved in induction of peroxisome proliferator-activated receptor γ (PPARγ), a representative anti-inflammatory transcription factor. As a crucial target of the SREBP2-EGR-1-PPARγ-associated signaling pathways, the mRNA stabilizer, human antigen R (HuR) was retained in nuclei, leading to reduced stability of pro-inflammatory chemokine transcripts. This mechanistic investigation provides clinical insights into protective roles of the epithelial cholesterol deficiency against excessive inflammatory responses via the SREBP2-HuR circuit, although the deficiency triggers transient pro-inflammatory signals.
Collapse
Affiliation(s)
- Seong-Hwan Park
- From the Laboratory of Mucosal Exposome and Biomodulation, Department of Biomedical Sciences and Medical Research Institute, Pusan National University School of Medicine, Yangsan 50612
| | - Juil Kim
- From the Laboratory of Mucosal Exposome and Biomodulation, Department of Biomedical Sciences and Medical Research Institute, Pusan National University School of Medicine, Yangsan 50612
| | - Mira Yu
- From the Laboratory of Mucosal Exposome and Biomodulation, Department of Biomedical Sciences and Medical Research Institute, Pusan National University School of Medicine, Yangsan 50612
| | - Jae-Hong Park
- the Department of Pediatrics, Pusan National University, Yangsan 50612
| | - Yong Sik Kim
- the Department of Pharmacology, College of Medicine, Seoul National University, Seoul 03080, and
| | - Yuseok Moon
- From the Laboratory of Mucosal Exposome and Biomodulation, Department of Biomedical Sciences and Medical Research Institute, Pusan National University School of Medicine, Yangsan 50612,; the Immunoregulatory Therapeutics Group in Brain Busan 21 Project, Busan 46241, Korea.
| |
Collapse
|
36
|
Higginson EE, Galen JE, Levine MM, Tennant SM. Microgravity as a biological tool to examine host-pathogen interactions and to guide development of therapeutics and preventatives that target pathogenic bacteria. Pathog Dis 2016; 74:ftw095. [PMID: 27630185 DOI: 10.1093/femspd/ftw095] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/31/2016] [Indexed: 12/16/2022] Open
Abstract
Space exploration programs have long been interested in the effects of spaceflight on biology. This research is important not only in its relevance to future deep space exploration, but also because it has allowed investigators to ask questions about how gravity impacts cell behavior here on Earth. In the 1980s, scientists designed and built the first rotating wall vessel, capable of mimicking the low shear environment found in space. This vessel has since been used to investigate growth of both microorganisms and human tissue cells in low shear modeled microgravity conditions. Bacterial behavior has been shown to be altered both in space and under simulated microgravity conditions. In some cases, bacteria appear attenuated, whereas in others virulence is enhanced. This has consequences not only for manned spaceflight, but poses larger questions about the ability of bacteria to sense the world around them. By using the microgravity environment as a tool, we can exploit this phenomenon in the search for new therapeutics and preventatives against pathogenic bacteria for use both in space and on Earth.
Collapse
Affiliation(s)
- Ellen E Higginson
- Center for Vaccine Development and Institute for Global Health, University of Maryland School of Medicine, Baltimore, MD 21201, USA Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - James E Galen
- Center for Vaccine Development and Institute for Global Health, University of Maryland School of Medicine, Baltimore, MD 21201, USA Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Myron M Levine
- Center for Vaccine Development and Institute for Global Health, University of Maryland School of Medicine, Baltimore, MD 21201, USA Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Sharon M Tennant
- Center for Vaccine Development and Institute for Global Health, University of Maryland School of Medicine, Baltimore, MD 21201, USA Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
37
|
Manjunatha UH, Chao AT, Leong FJ, Diagana TT. Cryptosporidiosis Drug Discovery: Opportunities and Challenges. ACS Infect Dis 2016; 2:530-7. [PMID: 27626293 DOI: 10.1021/acsinfecdis.6b00094] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The apicomplexan parasite Cryptosporidium is the second most important diarrheal pathogen causing life-threatening diarrhea in children, which is also associated with long-term growth faltering and cognitive deficiency. Cryptosporidiosis is a parasitic disease of public health concern caused by Cryptosporidium parvum and Cryptosporidium hominis. Currently, nitazoxanide is the only approved treatment for cryptosporidium infections. Unfortunately, it has limited efficacy in the most vulnerable patients, thus there is an urgent need for a safe and efficacious cryptosporidiosis drug. In this work, we present our current perspectives on the target product profile for novel cryptosporidiosis therapies and the perceived challenges and possible mitigation plans at different stages in the cryptosporidiosis drug discovery process.
Collapse
Affiliation(s)
- Ujjini H. Manjunatha
- Novartis Institute for Tropical Diseases, 10 Biopolis Road, #05-01, Singapore 138670
| | - Alexander T. Chao
- Novartis Institute for Tropical Diseases, 10 Biopolis Road, #05-01, Singapore 138670
| | - F. Joel Leong
- Novartis Institute for Tropical Diseases, 10 Biopolis Road, #05-01, Singapore 138670
| | - Thierry T. Diagana
- Novartis Institute for Tropical Diseases, 10 Biopolis Road, #05-01, Singapore 138670
| |
Collapse
|
38
|
Salerno-Goncalves R, Fasano A, Sztein MB. Development of a Multicellular Three-dimensional Organotypic Model of the Human Intestinal Mucosa Grown Under Microgravity. J Vis Exp 2016. [PMID: 27500889 DOI: 10.3791/54148] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Because cells growing in a three-dimensional (3-D) environment have the potential to bridge many gaps of cell cultivation in 2-D environments (e.g., flasks or dishes). In fact, it is widely recognized that cells grown in flasks or dishes tend to de-differentiate and lose specialized features of the tissues from which they were derived. Currently, there are mainly two types of 3-D culture systems where the cells are seeded into scaffolds mimicking the native extracellular matrix (ECM): (a) static models and (b) models using bioreactors. The first breakthrough was the static 3-D models. 3-D models using bioreactors such as the rotating-wall-vessel (RWV) bioreactors are a more recent development. The original concept of the RWV bioreactors was developed at NASA's Johnson Space Center in the early 1990s and is believed to overcome the limitations of static models such as the development of hypoxic, necrotic cores. The RWV bioreactors might circumvent this problem by providing fluid dynamics that allow the efficient diffusion of nutrients and oxygen. These bioreactors consist of a rotator base that serves to support and rotate two different formats of culture vessels that differ by their aeration source type: (1) Slow Turning Lateral Vessels (STLVs) with a co-axial oxygenator in the center, or (2) High Aspect Ratio Vessels (HARVs) with oxygenation via a flat, silicone rubber gas transfer membrane. These vessels allow efficient gas transfer while avoiding bubble formation and consequent turbulence. These conditions result in laminar flow and minimal shear force that models reduced gravity (microgravity) inside the culture vessel. Here we describe the development of a multicellular 3-D organotypic model of the human intestinal mucosa composed of an intestinal epithelial cell line and primary human lymphocytes, endothelial cells and fibroblasts cultured under microgravity provided by the RWV bioreactor.
Collapse
Affiliation(s)
| | - Alessio Fasano
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children
| | - Marcelo B Sztein
- Center for Vaccine Development, Department of Pediatrics, University of Maryland School of Medicine
| |
Collapse
|
39
|
Do KH, Park SH, Kim J, Yu M, Moon Y. Ribosome Inactivation Leads to Attenuation of Intestinal Polymeric Ig Receptor Expression via Differential Regulation of Human Antigen R. THE JOURNAL OF IMMUNOLOGY 2016; 197:847-58. [DOI: 10.4049/jimmunol.1502047] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 05/12/2016] [Indexed: 02/07/2023]
|
40
|
Korpe PS, Haque R, Gilchrist C, Valencia C, Niu F, Lu M, Ma JZ, Petri SE, Reichman D, Kabir M, Duggal P, Petri WA. Natural History of Cryptosporidiosis in a Longitudinal Study of Slum-Dwelling Bangladeshi Children: Association with Severe Malnutrition. PLoS Negl Trop Dis 2016; 10:e0004564. [PMID: 27144404 PMCID: PMC4856361 DOI: 10.1371/journal.pntd.0004564] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Accepted: 03/01/2016] [Indexed: 12/18/2022] Open
Abstract
Background Cryptosporidiosis is a common cause of infectious diarrhea in young children worldwide, and is a significant contributor to under-five mortality. Current treatment options are limited in young children. In this study, we describe the natural history of Cryptosporidium spp. infection in a birth cohort of children in Bangladesh and evaluate for association with malnutrition. Methodology/Principal Findings This is a longitudinal birth cohort study of 392 slum-dwelling Bangladeshi children followed over the first two years of life from 2008 to 2014. Children were monitored for diarrheal disease, and stool was tested for intestinal protozoa. Anthropometric measurements were taken at 3-month intervals. A subset of Cryptosporidium positive stools were genotyped for species and revealed that C. hominis was isolated from over 90% of samples. In the first two years of life, 77% of children experienced at least one infection with Cryptosporidium spp. Non-diarrheal infection (67%) was more common than diarrheal infection (6.3%) although 27% of children had both types of infection. Extreme poverty was associated with higher rates of infection (chi-square, 49.7% vs 33.3%, p = 0.006). Malnutrition was common in this cohort, 56% of children had stunted growth by age two. Children with Cryptosporidium spp. infection had a greater than 2-fold increased risk of severe stunting at age two compared to uninfected children (odds ratio 2.69, 95% CI 1.17, 6.15, p = 0.019) independent of sex, income, maternal body-mass index, maternal education and weight for age adjusted z (WAZ) score at birth. Conclusions/Significance Cryptosporidium infection is common (77%) in this cohort of slum-dwelling Bangladeshi children, and both non-diarrheal and diarrheal infections are significantly associated with a child’s growth at 2 years of age. Diarrheal disease is a leading cause of death in young children worldwide. Cryptosporidium species are responsible for a large proportion of global burden of diarrhea. This study describes the natural history of cryptosporidiosis in a birth cohort of impoverished Bangladeshi children. Children were enrolled at birth and monitored for diarrhea twice a week for two years. Stool samples were tested for enteric protozoa. Children in this cohort had significant rates of malnutrition compared to the W.H.O. reference population, and extreme poverty was common. A majority of children were infected with Cryptosporidium spp, and we found that children who had at least one infection with Cryptosporidium spp during the two year follow up period were significantly more likely to have growth faltering by age 24 months. Cryptosporidiosis is a common infection in this cohort, and is associated with poverty and reduced growth during the first two years of life.
Collapse
Affiliation(s)
- Poonum S. Korpe
- Department of Epidemiology, Johns Hopkins School of Public Health, Baltimore, Maryland, United States of America
- * E-mail:
| | - Rashidul Haque
- International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Carol Gilchrist
- Department of Medicine, Division of Infectious Diseases, University of Virginia, Charlottesville, Virginia, United States of America
| | - Cristian Valencia
- Department of Epidemiology, Johns Hopkins School of Public Health, Baltimore, Maryland, United States of America
| | - Feiyang Niu
- Department of Statistics, University of Virginia, Charlottesville, Virginia, United States of America
| | - Miao Lu
- Department of Statistics, University of Virginia, Charlottesville, Virginia, United States of America
| | - Jennie Z. Ma
- Division of Biostatistics, Department of Public Health Sciences, University of Virginia, Charlottesville, Virginia, United States of America
| | - Sarah E. Petri
- Department of Animal and Veterinary Sciences, Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts, United States of America
| | - Daniel Reichman
- Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Mamun Kabir
- International Centre for Diarrhoeal Disease Research, Bangladesh, Dhaka, Bangladesh
| | - Priya Duggal
- Department of Epidemiology, Johns Hopkins School of Public Health, Baltimore, Maryland, United States of America
| | - William A. Petri
- Department of Medicine, Division of Infectious Diseases, University of Virginia, Charlottesville, Virginia, United States of America
| |
Collapse
|
41
|
Miyamoto Y, Eckmann L. Drug Development Against the Major Diarrhea-Causing Parasites of the Small Intestine, Cryptosporidium and Giardia. Front Microbiol 2015; 6:1208. [PMID: 26635732 PMCID: PMC4652082 DOI: 10.3389/fmicb.2015.01208] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 10/16/2015] [Indexed: 12/23/2022] Open
Abstract
Diarrheal diseases are among the leading causes of morbidity and mortality in the world, particularly among young children. A limited number of infectious agents account for most of these illnesses, raising the hope that advances in the treatment and prevention of these infections can have global health impact. The two most important parasitic causes of diarrheal disease are Cryptosporidium and Giardia. Both parasites infect predominantly the small intestine and colonize the lumen and epithelial surface, but do not invade deeper mucosal layers. This review discusses the therapeutic challenges, current treatment options, and drug development efforts against cryptosporidiosis and giardiasis. The goals of drug development against Cryptosporidium and Giardia are different. For Cryptosporidium, only one moderately effective drug (nitazoxanide) is available, so novel classes of more effective drugs are a high priority. Furthermore, new genetic technology to identify potential drug targets and better assays for functional evaluation of these targets throughout the parasite life cycle are needed for advancing anticryptosporidial drug design. By comparison, for Giardia, several classes of drugs with good efficacy exist, but dosing regimens are suboptimal and emerging resistance begins to threaten clinical utility. Consequently, improvements in potency and dosing, and the ability to overcome existing and prevent new forms of drug resistance are priorities in antigiardial drug development. Current work on new drugs against both infections has revealed promising strategies and new drug leads. However, the primary challenge for further drug development is the underlying economics, as both parasitic infections are considered Neglected Diseases with low funding priority and limited commercial interest. If a new urgency in medical progress against these infections can be raised at national funding agencies or philanthropic organizations, meaningful and timely progress is possible in treating and possibly preventing cryptosporidiosis and giardiasis.
Collapse
Affiliation(s)
- Yukiko Miyamoto
- Department of Medicine, University of California at San Diego, La Jolla CA, USA
| | - Lars Eckmann
- Department of Medicine, University of California at San Diego, La Jolla CA, USA
| |
Collapse
|
42
|
Morada M, Lee S, Gunther-Cummins L, Weiss LM, Widmer G, Tzipori S, Yarlett N. Continuous culture of Cryptosporidium parvum using hollow fiber technology. Int J Parasitol 2015; 46:21-9. [PMID: 26341006 DOI: 10.1016/j.ijpara.2015.07.006] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 07/23/2015] [Accepted: 07/24/2015] [Indexed: 02/08/2023]
Abstract
Diarrheal disease is a leading cause of pediatric death in economically low resource countries. Cryptosporidium spp. are the second largest member of this group and the only member for which no treatment exists. One of the handicaps to developing chemotherapy is the lack of a reproducible long-term culture method permitting in vitro drug screening beyond 48 h. We have adapted the well-established hollow fiber technology to provide an environment that mimics the gut by delivering nutrients and oxygen from the basal layer upwards while allowing separate redox and nutrient control of the lumen for parasite development. Using this technique, oocyst production was maintained for >6 months, producing approximately 1×10(8)oocysts ml(-1)day(-1), compared with 48 h with a yield of 1×10(6)oocysts ml(-1) in two-dimensional cultures. Oocysts, after 4 and 20 weeks in culture, produced a chronic infection in a TCR-α-deficient mouse model. In vivo infectivity of oocysts was confirmed using oocysts from a 6 week culture in a dexamethasone immunosuppressed mouse model.
Collapse
Affiliation(s)
- Mary Morada
- Haskins Laboratories, and Department of Chemistry and Physical Sciences, Pace University, New York, USA
| | - Sangun Lee
- Cummings School of Veterinary Medicine, Tufts University, N. Grafton, MA, USA
| | | | - Louis M Weiss
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, USA; Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Giovanni Widmer
- Cummings School of Veterinary Medicine, Tufts University, N. Grafton, MA, USA
| | - Saul Tzipori
- Cummings School of Veterinary Medicine, Tufts University, N. Grafton, MA, USA
| | - Nigel Yarlett
- Haskins Laboratories, and Department of Chemistry and Physical Sciences, Pace University, New York, USA.
| |
Collapse
|
43
|
The Global Burden of Pediatric Cryptosporidium Infections. CURRENT TROPICAL MEDICINE REPORTS 2015. [DOI: 10.1007/s40475-015-0053-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
44
|
Choi HJ, Kim HG, Kim J, Park SH, Park J, Oh CG, Do KH, Lee SJ, Park YC, Ahn SC, Kim YS, Moon Y. Pro-apoptotic action of macrophage inhibitory cytokine 1 and counteraction of activating transcription factor 3 in carrageenan-exposed enterocytes. Toxicol Lett 2014; 231:1-8. [PMID: 25180886 DOI: 10.1016/j.toxlet.2014.08.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 08/28/2014] [Accepted: 08/29/2014] [Indexed: 12/15/2022]
Abstract
Carrageenan (CGN), a widely used food additive, has been shown to injure the epithelial barrier in animal models. This type of damage is a clinical feature of inflammatory bowel disease (IBD) in humans. In the present study, the effects of CGN on pro-apoptotic responses associated with macrophage inhibitory cytokine 1 (MIC-1) regulation in human enterocytes were evaluated. CGN up-regulated the expression of MIC-1 that promoted epithelial cell apoptosis. Although MIC-1 induction was dependent on pro-apoptotic p53 protein, the pro-survival protein activating transcription factor 3 (ATF3) was negatively regulated by p53 expression. However, MIC-1 enhanced the expression of the pro-survival protein ATF3 in enterocytes exposed to CGN. Functionally, MIC-1-mediated epithelial cell apoptosis was counteracted by the pro-survival action of ATF3 in response to CGN exposure. These findings demonstrated that the counterbalance between MIC-1 and ATF3 is critical for deciding the fate of enterocytes under the food chemical stress.
Collapse
Affiliation(s)
- Hye Jin Choi
- Laboratory of Mucosal Exposome and Biomodulation, Department of Biomedical Sciences, Pusan National University School of Medicine, Yangsan, South Korea
| | - Hwi-Gon Kim
- Department of Obstetrics and Gynecology, Medical Research Institute, Pusan National University, Pusan, South Korea
| | - Juil Kim
- Laboratory of Mucosal Exposome and Biomodulation, Department of Biomedical Sciences, Pusan National University School of Medicine, Yangsan, South Korea
| | - Seong-Hwan Park
- Laboratory of Mucosal Exposome and Biomodulation, Department of Biomedical Sciences, Pusan National University School of Medicine, Yangsan, South Korea
| | - Jiyeon Park
- Laboratory of Mucosal Exposome and Biomodulation, Department of Biomedical Sciences, Pusan National University School of Medicine, Yangsan, South Korea
| | - Chang Gyu Oh
- Laboratory of Mucosal Exposome and Biomodulation, Department of Biomedical Sciences, Pusan National University School of Medicine, Yangsan, South Korea
| | - Kee Hun Do
- Laboratory of Mucosal Exposome and Biomodulation, Department of Biomedical Sciences, Pusan National University School of Medicine, Yangsan, South Korea
| | - Seung Joon Lee
- Laboratory of Mucosal Exposome and Biomodulation, Department of Biomedical Sciences, Pusan National University School of Medicine, Yangsan, South Korea
| | - Young Chul Park
- Department of Microbiology and Immunology and Medical Research Institute, Pusan National University, Pusan, South Korea
| | - Soon Cheol Ahn
- Department of Microbiology and Immunology and Medical Research Institute, Pusan National University, Pusan, South Korea
| | - Yong Sik Kim
- Department of Pharmacology, Seoul National University College of Medicine, Seoul, South Korea
| | - Yuseok Moon
- Laboratory of Mucosal Exposome and Biomodulation, Department of Biomedical Sciences, Pusan National University School of Medicine, Yangsan, South Korea; Department of Microbiology and Immunology and Medical Research Institute, Pusan National University, Pusan, South Korea; Immunoregulatory Therapeutics Group in Brain Busan 21 Project, Busan, South Korea.
| |
Collapse
|
45
|
David J, Sayer NM, Sarkar-Tyson M. The use of a three-dimensional cell culture model to investigate host-pathogen interactions of Francisella tularensis in human lung epithelial cells. Microbes Infect 2014; 16:735-45. [PMID: 24796635 DOI: 10.1016/j.micinf.2014.04.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Revised: 01/14/2014] [Accepted: 04/07/2014] [Indexed: 01/25/2023]
Abstract
Francisella tularensis inhalation results in bacterial interaction with numerous lung cell types, including those of the epithelium. This work investigates a three-dimensional cell-culture system to characterise the epithelial response to F. tularensis. Immortalised human pneumocytes (A549s) grown in rotating-wall vessel (RWV) bioreactors display an in vivo-like phenotype, which has been confirmed to be driven by specific transcriptional events (8454 genes, p ≤ 0.05). These data support the RWV model as a more in vivo-like culture system to investigate the lung epithelium, compared to monolayer counterparts. RWV-cultured A549s were infected with F. tularensis SchuS4 and LVS and intracellular replication mapped over 22 h compared to monolayer cells. The RWV-cultured A549s were more resistant to SchuS4 and LVS infection (p ≤ 0.05). Transcriptomics identified 2086 genes (p ≤ 0.05) as candidates for host-pathogen interactions which result in the observed increase in resistance of the RWV-cultured A549 cells. Gene and pathway analysis identified processes involved in MMP modulation, endocytosis, mucin production and the complement pathway amongst others. The role of these pathways during infection was further characterised using chemical inhibitors. This work has revealed several new hypotheses worthy of further testing in order to understand the epithelial host response to F. tularensis infection.
Collapse
Affiliation(s)
- Jonathan David
- Biomedical Sciences, Dstl, Porton Down, Salisbury, Wiltshire SP4 0JQ, UK.
| | - Natalie M Sayer
- Biomedical Sciences, Dstl, Porton Down, Salisbury, Wiltshire SP4 0JQ, UK
| | - Mitali Sarkar-Tyson
- Biomedical Sciences, Dstl, Porton Down, Salisbury, Wiltshire SP4 0JQ, UK; School of Pathology and Laboratory Medicine, University of Western Australia, QEII Medical Centre, Monash Avenue, Nedlands, WA 6009, USA
| |
Collapse
|
46
|
Park SH, Do KH, Choi HJ, Kim J, Kim KH, Park J, Oh CG, Moon Y. Novel regulatory action of ribosomal inactivation on epithelial Nod2-linked proinflammatory signals in two convergent ATF3-associated pathways. THE JOURNAL OF IMMUNOLOGY 2013; 191:5170-81. [PMID: 24098051 DOI: 10.4049/jimmunol.1301145] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
In response to excessive nucleotide-binding oligomerization domain-containing protein 2 (Nod2) stimulation caused by mucosal bacterial components, gut epithelia need to activate regulatory machinery to maintain epithelial homeostasis. Activating transcription factor 3 (ATF3) is a representative regulator in the negative feedback loop that modulates TLR-associated inflammatory responses. In the current study, the regulatory effects of ribosomal stress-induced ATF3 on Nod2-stimulated proinflammatory signals were assessed. Ribosomal inactivation caused persistent ATF3 expression that in turn suppressed proinflammatory chemokine production facilitated by Nod2. Decreased chemokine production was due to attenuation of Nod2-activated NF-κB and early growth response protein 1 (EGR-1) signals by ATF3. However, the underlying molecular mechanisms involve two convergent regulatory pathways. Although ATF3 induced by ribosomal inactivation regulated Nod2-induced EGR-1 expression epigenetically through the recruitment of histone deacetylase 1, NF-κB regulation was associated with posttranscriptional regulation by ATF3 rather than epigenetic modification. ATF3 induced by ribosomal inactivation led to the destabilization of p65 mRNA caused by nuclear entrapment of transcript-stabilizing human Ag R protein via direct interaction with ATF3. These findings demonstrate that ribosomal stress-induced ATF3 is a critical regulator in the convergent pathways between EGR-1 and NF-κB, which contributes to the suppression of Nod2-activated proinflammatory gene expression.
Collapse
Affiliation(s)
- Seong-Hwan Park
- Laboratory of Mucosal Exposome and Biomodulation, Department of Microbiology and Immunology, Medical Research Institute, Pusan National University School of Medicine, Yangsan 626-870, Korea
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Do KH, Choi HJ, Kim J, Park SH, Kim KH, Moon Y. SOCS3 Regulates BAFF in Human Enterocytes under Ribosomal Stress. THE JOURNAL OF IMMUNOLOGY 2013; 190:6501-10. [DOI: 10.4049/jimmunol.1203004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
48
|
Al-Mathal EM, Alsalem AA. Pomegranate (Punica granatum) peel is effective in a murine model of experimental Cryptosporidium parvum ultrastructural studies of the ileum. Exp Parasitol 2013; 134:482-94. [PMID: 23684569 DOI: 10.1016/j.exppara.2013.05.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2012] [Revised: 03/27/2013] [Accepted: 05/02/2013] [Indexed: 11/24/2022]
Abstract
The current treatments for cryptosporidiosis are ineffective, and there is an urgent need to search for more effective and safer alternatives. One such alternative may be treatments derived from natural resources. The pomegranate peel has been used effectively in traditional medicine to cure diarrhea and dysentery. The purpose of this study was to examine the effectiveness of a Punica granatum (pomegranate) peel suspension as a treatment for Cryptosporidium parvum infection. In this study, the effects of this treatment on the ultrastructure of both the intestinal epithelial layer of infected nursling mice and the parasite were observed with a transmission electron microscope. The histological study focused on the examination of the microvilli, columnar epithelium, goblet cells, lamina propria, and crypts of Lieberkuhn. Examination of the ileums of infected mice that received the pomegranate peel suspension demonstrated that the general structure of the ileal tissue of these mice was similar to that of the control group. In the infected mice treated with the suspension, but not the infected/untreated mice, there was an improvement in all ultrastructure aspects at 28days post-inoculation. The study of the ultrastructure of the parasite (C. parvum) in mice treated with the suspension showed that there was decomposition in the parasite to the extent that in some cases we were unable to identify the stage of the parasite due to the severe degeneration. Significant decomposition of the nutrition organ was also observed. Additionally, microgamonte and macrogamonte were not observed in the suspension-treated group, explaining the disappearance of the sexual phases of the parasite in the lumens of this group. In all, this examination demonstrated the restoration of the normal structures of villi and the disappearance of acute symptoms in the suspension-treated mice and showed that the suspension directly affected the parasite at various stages of its development and led to its decomposition and death.
Collapse
Affiliation(s)
- Ebtisam M Al-Mathal
- Department of Biology, College of Science, University of Dammam, Dammam 31311, Saudi Arabia.
| | | |
Collapse
|
49
|
Liévin-Le Moal V. Dysfunctions at human intestinal barrier by water-borne protozoan parasites: lessons from cultured human fully differentiated colon cancer cell lines. Cell Microbiol 2013; 15:860-9. [PMID: 23437821 DOI: 10.1111/cmi.12126] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2012] [Revised: 02/04/2013] [Accepted: 02/06/2013] [Indexed: 12/21/2022]
Abstract
Some water-borne protozoan parasites induce diseases through their membrane-associated functional structures and virulence factors that hijack the host cellular molecules and signalling pathways leading to structural and functional lesions in the intestinal barrier. In this Microreview we analyse the insights on the mechanisms of pathogenesis of Entamoeba intestinalis, Giardia and Cryptosporidium observed in the human colon carcinoma fully differentiated colon cancer cell lines, cell subpopulations and clones expressing the structural and functional characteristics of highly specialized fully differentiated epithelial cells lining the intestinal epithelium and mimicking structurally and functionally an intestinal barrier.
Collapse
Affiliation(s)
- Vanessa Liévin-Le Moal
- CNRS, UMR 8076 BioCIS, Team Antiparasitic chemotherapy, Faculty of Pharmacy, Châtenay-Malabry, 92296, France.
| |
Collapse
|
50
|
Prolonged NF-κB activation by a macrophage inhibitory cytokine 1-linked signal in enteropathogenic Escherichia coli-infected epithelial cells. Infect Immun 2013; 81:1860-9. [PMID: 23403560 DOI: 10.1128/iai.00162-13] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Intestinal epithelial activation of nuclear factor kappa B (NF-κB) exerts both detrimental and beneficial functions in response to various luminal insults, including ones associated with mucosa-associated pathogens. Gastrointestinal infection with enteropathogenic Escherichia coli (EPEC) causes severe injuries in epithelial integrity and leads to watery diarrhea. The present study was conducted to investigate the prolonged epithelial responses to persistent EPEC infection via NF-κB activation. EPEC infection led to sustained activation of NF-κB signal in mouse intestinal epithelial cells in vivo and in vitro, which was positively associated with a type III secretion system, whereas early NF-κB is regulated. Moreover, prolonged NF-κB activation was found to be a part of macrophage inhibitory cytokine 1 (MIC-1)-mediated signaling activation, a novel link between NF-κB signaling and infection-associated epithelial stress. EPEC infection induced gene expression of MIC-1, a member of the transforming growth factor β (TGF-β) superfamily, which then activated TGF-β-activated kinase 1 and consequently led to NF-κB activation. Functionally, both EPEC-induced MIC-1 and NF-κB signaling mediated epithelial survival by enhancing the expression of cyclin D1, a target of NF-κB. In summary, the results of the present study suggest that MIC-1 serves as a mediator of prolonged NF-κB activation, which is critical in maintaining gut epithelial integrity in response to infection-induced injuries.
Collapse
|